EP2301967A2 - Single domain antibodies for nasal administration - Google Patents

Single domain antibodies for nasal administration Download PDF

Info

Publication number
EP2301967A2
EP2301967A2 EP10178270A EP10178270A EP2301967A2 EP 2301967 A2 EP2301967 A2 EP 2301967A2 EP 10178270 A EP10178270 A EP 10178270A EP 10178270 A EP10178270 A EP 10178270A EP 2301967 A2 EP2301967 A2 EP 2301967A2
Authority
EP
European Patent Office
Prior art keywords
polypeptide construct
single domain
vhh
target
directed against
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP10178270A
Other languages
German (de)
French (fr)
Other versions
EP2301967A3 (en
Inventor
Karen Silence
Mark Vaeck
Paul P.M.P. Van Bergen En Henegouwen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ablynx NV
Original Assignee
Ablynx NV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ablynx NV filed Critical Ablynx NV
Priority to EP10178270A priority Critical patent/EP2301967A3/en
Priority claimed from EP03775004A external-priority patent/EP1558650A2/en
Publication of EP2301967A2 publication Critical patent/EP2301967A2/en
Publication of EP2301967A3 publication Critical patent/EP2301967A3/en
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/249Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/36Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/626Diabody or triabody
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • Polypeptide therapeutics and in particular antibody-based therapeutics have significant potential as drugs because they have extraordinarily specificity to their target and a low inherent toxicity. However, they have one important drawback: these are complex, large molecules and therefore relatively unstable, and they are sensitive to breakdown by proteases. Because the degradation they undergo during passage through, for instance, the gastrointestinal tract, administration of conventional antibodies and their derived fragments or single-chain formats (e.g. scFv's) is not very effective. This means that conventional antibody drugs cannot be administered orally, sublingually, topically, nasally, vaginally, rectally or by inhalation because they are not resistant to the low pH at these sites, the action of proteases at these sites and in the blood and/or because of their large size.
  • scFv's single-chain formats
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is antigen of MMP and the disorder is cancer.
  • such lyophilized VHH formulations can be reconstituted with a diluent to generate a stable reconstituted formulation suitable for subcutaneous administration.
  • anti-lgE antibody formulations (Example 1; US 6267958 , EP 841946 ) have been prepared which are useful for treating allergic asthma.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the nose, upper respiratory tract and/or lung without being inactivated, by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the tissues beneath the tongue without being inactivated, by administering orally to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • a colon cancer specific antigen is epidermal growth factor receptor (EGFR) which is an essential mediator of cell division in mammalian cells and is a recognised cellular oncogene. After the binding of EGF to its receptor (EGFR), a signaling cascade is initiated resulting in cell development.
  • EGFR epidermal growth factor receptor
  • the EGFR is also involved in human tumorigenesis as it is overexpressed on cells associated with epithelial malignancies located in sites such as the head, neck, lung, colon.
  • Another aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against EGFR for use in the treatment, prevention and/or alleviation of disorders relating to EGFR-mediated cancer, wherein said VHH is administered orally, sublingually, topically, nasally, intravenously, subcutaneously, vaginally, rectally or by inhalation (Examples 25 to 31).
  • Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to EGFR-mediated cancer, comprising administering to a subject a polypeptide construct comprising at least one single domain antibody directed against EGFR orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation.
  • a polypeptide construct of the invention comprises at least one single domain antibody directed against IgE, said single domain antibody corresponding to a sequence represented by any of SEX ID NOs: 1 to 11. Said sequences are anti-IgE Camelidae VHHs.
  • the invention also relates to a method of identifying single domain antibodies (e.g. VHHs) harbouring specific sequences which facilitates the delivery or transport of the anti-target single domain antibodies across human or animal tissues (as described in US 6361938 ), including without limitation GIT epithelial layers, alveolar cells, endothelial of the blood-brain barrier, vascular smooth muscle cells, vascular endothelial cells, renal epithelial cells, M cells of the Peyers Patch, and hepatocytes.
  • delivery systems could be used in conjunction with the VHH's of the invention, comprising nanoparticles, microparticles, liposomes, micelles, cyclodextrines.
  • One embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies specific for an internalising cellular receptor as disclosed herein, further comprising an antigen.
  • One embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies specific for an internalising cellular receptor as disclosed herein, wherein said receptor is EGFR.
  • sequestration In general internalization of receptors occurs upon binding of the agonistic ligand in a proces called sequestration. In order to ensure that extracellular signals are translated into intracellular signals of appropriate magnitude and specificity, the signalling cascades are tightly regulated via the process of sequestration, whereby receptors are physically removed from the cell surface by internalization to a cytosolic compartment ( Carman, C.V. and Benovic, J.L. Current Opinion in Neurobiology 1998, 8: 335-344 ). This implies that only agonistic ligands or antibodies indeed are expected to internalize via such receptors. In terms of therapeutic use it is not a desired effect that the antibody first triggers proliferation of the tumorcells, before it can deliver a toxic payload to the interiour of the cell.
  • formulation technology may be applied to release a maximum amount of VHHs in the right location (in the stomach, in the colon, etc.). This method of delivery is important for treating, prevent and/or alleviate the symptoms of disorder whose targets that are located in the gut system.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the gut system without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the vaginal and/or rectal tract without being inactivated.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic delivered to the nose, upper respiratory tract and lung, by administering to a subject a polypeptide construct as disclosed herein wherein one or more single domain antibodies are specific for an antigen related to the disorder, by inhalation through the mouth or nose.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the nose, upper respiratory tract and/or lung without being inactivated, by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and devices.
  • Example 7 Efficacy in an animal model for IBD
  • the library was grown at 37°C in 10 ml 2xTY medium containing 2% glucose, and 100 ⁇ g/ml ampicillin, until the OD 600nm reached 0.5.
  • M13K07 phages (10 12 ) were added and the mixture was incubated at 37°C for 2 x 30 minutes, first without shaking, then with shaking at 100 rpm. Cells were centrifuged for 5 minutes at 4,500 rpm at room temperature. The bacterial pellet was resuspended in 50 ml of 2xTY medium containing 100 ⁇ g/ml ampicillin and 25 ⁇ g/ml kanamycin, and incubated overnight at 37°C with vigorously shaking at 250 rpm.
  • Microtiter wells were coated with human IFN-y at different concentrations of 10-0.4 ⁇ g/well overnight at 4°C. Plates were washed 5 times with PBS/0.05%Tween-20. Wells were blocked with PBS)+ 1% caseine for 2 hrs at room temperature. Phages were incubated for 2 hrs at room temperature. Wells were washed 20 times with PBS+0.05%Tween-20. The two final washes were performed using PBS. Specific phages were eluted using 1 to 2 ⁇ g of IFN-y R1 (R&D Systems, Cat Nr: 673-IR/CF) for 1 hr.
  • IFN-y R1 R&D Systems, Cat Nr: 673-IR/CF
  • FIG. 11 shows that EGFRllla42 is able to internalize Her-14 (panel A) but not 3T3 cells (panel B). Subsequently, FACS analysis demonstrated that nanobody EGFR-llla42 is able to bind both A431 and Her-1 4, but not 3T3 (data not shown).
  • PNPP p-nitrophenyl-phosphate, 2 mg/ml in 1 M diethanolamine, 1mM Mg 2 SO 4 , pH9.8 and the signals are measured after 30 minutes at 405 nm.
  • Table 18 An example of the expected number of positive clones versus the number of clones tested in ELISA for each selection is presented in Table 18.
  • Table 5 Second round selection using neutravidine coated tubes as described in Example 1 2 nM lgE 2 nM lgE 0 nM lgE 0 nM lgE Elution buffy coat cells Elution Lysozyme Elution buffy coat cells Elution Lysozyme Llama 2 (selection 5 ⁇ g/ ml lgE: 400 x enrichment) 1.5 10 8 1.5 10 7 3 10 5 3 10 5 Enrichment compared to lysozyme elution 10 x Llama 4 (selection 5 ⁇ g/ ml lgE: 140 x enrichment) 3.3 10 7 2.2 10 7 3 10 3 6 10 3 Enrichment compared to lysozyme elation 1.5 x Table 6: Number of clones that score positive for binding to both human lgE and chimeric lgE versus the number of clones tested in ELISA as described in Example 1 Selection with 5 ⁇ g/ml Selection with

Abstract

The invention relates to a method suitable for administering protein therapeutic molecules orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation so as to avoid inactivation, by using VHH polypeptides derived from Camelidae antibodies. The invention further relates to the said therapeutic molecules. The invention further a method for delivering therapeutic molecules to the interior of cells. The invention furthe relates to anti-lgE therapeutic molecules.

Description

    BACKGROUND
  • Polypeptide therapeutics and in particular antibody-based therapeutics have significant potential as drugs because they have exquisite specificity to their target and a low inherent toxicity. However, they have one important drawback: these are complex, large molecules and therefore relatively unstable, and they are sensitive to breakdown by proteases. Because the degradation they undergo during passage through, for instance, the gastrointestinal tract, administration of conventional antibodies and their derived fragments or single-chain formats (e.g. scFv's) is not very effective. This means that conventional antibody drugs cannot be administered orally, sublingually, topically, nasally, vaginally, rectally or by inhalation because they are not resistant to the low pH at these sites, the action of proteases at these sites and in the blood and/or because of their large size. They have to be administered by injection (intravenously, subcutaneously, etc.) to overcome some of these problems. Administration by injection is therefore the most frequently used method of administration although the method has many disadvantages, for example: (a) poor tolerance by patients, especially when treating chronic disorder (b) a consequent risk of poor compliance with the dosage when the drug is not a 'life saver'; (c) difficulty of carrying out self-administration by the patient; (d) possible non-availability of suitable surroundings for carrying out the procedure in an aseptic manner; (e) requires specialist training in order to use a hypodermic syringe or needle correctly and safely. A method for the delivery of therapeutic polypeptides which avoids the need for injection has not only cost/time savings, but would also be more convenient and more comfortable for the subject.
  • In most animal cells, a specialised pathway is present for uptake of specific macromolecules from the extracellular fluid. The macromolecules that bind to specific cell-surface receptors are internalized, a process called receptor-mediated endocytosis. Receptor internalization is based on the principle of regulation of signal transduction by a process called sequestration, whereby bound agonistic (i.e. receptor activation) ligands are recovered from the cell surface in complex with the receptor. For many applications it is necessary to deliver effector molecules across the cell membrane and into the cytosol. This can be achieved by taking advantage of such internalizing receptors. Antibodies have been described that internalize upon binding to internalizing receptors. However, they have important drawbacks: these antibodies are complex, large molecules and therefore relatively unstable, and they are sensitive to breakdown by proteases. Moreover, the domains of such antibodies are held together by disulphide bonds that dissociate in the reducing environment of the cytoplasm leading to a substantial loss of binding activity. Therefore, they cannot be used to target intracellular proteins.
  • Another process that relies on internalisation is the efficient induction of an immune response. In particular, a T-cell response depends heavily on efficient presentation of certain epitopes to the T cells by antigen presenting cells (APCs). In the case of a protein antigen this means that the APC has to take up the protein, internally process it (this is cleaving it) and express certain peptide fragments on its surface in association with MHC (major histocompatibility complex) or HLA molecules. One major and critical event in this process is the efficient uptake of the protein antigen by its APC. Techniques which can enhance antigen uptake by APCs enables an immune response to be elicited against antigens which naturally elicit a weak or no immune response. Therefore, a technique which can boost an immune response against antigenic antigens, naturally weak or non-immunogenic antigens has important implications for vaccination programs.
  • IgE plays a major role in allergic disease by causing the release of histamine and other inflammatory mediatord from mast cells. A mainstay of treatment of allergic disease, including asthma, is allergen avoidance and treatment of symptoms. Presently, the most effective treatments of allergic diseases are directed towards a regulation of the inflammatory process with corticosteroids. A more direct approach without the negative effects of corticosteroids consists in regulating the allergic process at the level of the initiatior of the allergic inflammation, IgE, via an anti-IgE.
    The concept of using anti-IgE antibodies as a treatment for allergy has been widely disclosed in the scientific literature. A few representative examples are as follows. Baniyash and Eshhar (European Journal of Immunology 14:799-807 (1984)) demonstrated that an anti-IgE monoclonal antibody could specifically block passive cutaneous anaphylaxis reaction when injected intradermally before challenging with the antigen; U.S. 4,714,759 discloses a product and process for treating allergy, using an antibody specific for IgE: and Rup and Kahn (International Archives Allergy and Applied Immunology, 89:387-393 (1989) discuss the prevention of the development of allergic responses with monoclonal antibodies which block mast cell-lgE sensitization.
    Anti-IgE antibodies which block the binding of IgE to its receptor on basophils and which fail to bind to IgE bound to the receptor, thereby avoiding histamine release are disclosed, for example, by Rup and Kahn (supra), by Baniyash et al. (Molecular Immunology 25:705-711. 1988), and by Hook et al. (Federation of American Societies for Experimental Biology, 71st Annual Meeting, Abstract #6008, 1987).
    Antagonists of IgE in the form of receptors, anti-IgE antibodies, binding factors, or fragments thereof have been disclosed in the art. For example, U.S. 4,962,035 discloses DNA encoding the alpha-subunit of the mast cell igE receptor or an IgE binding fragment thereof. Hook et al. (Federation Proceedings Vol. 40, No. 3, Abstract #4177) disclose monoclonal antibodies, of which one type is anti-idiotypic, a second type binds to common IgE determinants, and a third type is directed towards determinants hidden when IgE is on the basophil surface.
    U.S. 4,940,782 discloses monoclonal antibodies which react with free IgE and thereby inhibit IgE binding to mast cells, and react with IgE when it is bound to the B-cell FcE receptor, but do not bind with IgE when it is bound to the mast cell FcE receptor, nor block the binding of IgE to the B-cell receptor.
    U.S. 4,946,788 discloses a purified IgE binding factor and fragments thereof, and monoclonal antibodies which react with IgE binding factor and lymphocyte cellular receptors for IgE, and derivatives thereof.
    U.S. 5,091,313 discloses antigenic epitopes associated with the extracellular segment of the domain which anchors immunoglobulins to the B cell membrane. The epitopes recognized are present on IgE-bearing B cells but not basophils or in the secreted, soluble form of IgE. U.S. 5,252,467 discloses a method for producing antibodies specific for such antigenic epitopes. U.S. 5,231,026 discloses DNA encoding murine-human antibodies specific for such antigenic epitopes.
    U.S. 4,714,759 discloses an immunotoxin in the form of an antibody or an antibody fragment coupled to a toxin to treat allergy.
    Presta et al. (J. Immunol. 151:2623-2632 (1993)) disclose a humanized anti-IgE antibody that prevents the binding of free IgE to FceRl but does not bind to Fc□Rl-bound IgE. Copending WO93/0417 discloses polypeptides which bind differentially to the high- and low-affinity IgE receptors.
    U.S. 5,428,133 discloses anti-IgE antibodies as a therapy for allergy, especially antibodies which bind to IgE on B cells, but not IgE on basophils. This publication mentions the possibility of treating asthma with such antibodies. U.S. 5,422,258 discloses a method for making such antibodies.
    EP0841946 discloses methods for treating allergic asthma using IgE antagonists.
  • AIMS OF THE INVENTION
  • The aim of the invention is to provide a method of administering protein therapeutic molecules orally, sublingually, topically, nasally, vaginally, rectally, intraveneously, subcutaneously or by inhalation which overcomes the problems of the prior art. It is a further aim to provide said therapeutic molecules.
  • Another aim of the invention is to provide a method for delivering therapeutic substances to the interior of cells via internalizing receptors without receptor activation.
  • It is further aim of the invention to provide a therapeutic agent for the treatment of allergies.
  • It is a further aim of the invention to provide therapeutic nanobodies.
  • SUMMARY OF THE INVENTION
  • One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against IgE.
  • Another embodiment of the present invention is a polypeptide construct as described above wherein at least one single domain antibody is a Camelidae VHH.
  • Another embodiment of the present invention is a polypeptide construct as described above wherein at least one single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 11.
  • Another embodiment of the present invention is a polypeptide construct as described above, wherein the number of anti-IgE single domain antibodies is at least two.
  • Another embodiment of the present invention is a polypeptide construct as described above, wherein at least one single domain antibody is a humanized Camelidae VHH.
  • Another embodiment of the present invention is a polypeptide construct as described above, wherein a single domain antibody is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length single domain antibody.
  • Another embodiment of the present invention is a polypeptide construct as described above, wherein the polypeptide construct is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length polypeptide construct.
  • Another embodiment of the present invention is a nucleic acid encoding a polypeptide construct as described above.
  • Another embodiment of the present invention is a polypeptide construct as described above for treating and/or preventing and/or alleviating disorders relating to inflammatory processes.
  • Another embodiment of the present invention is a use of a polypeptide construct as described above for the preparation of a medicament for treating and/or preventing and/or alleviating disorders relating to inflammatory reactions.
  • Another embodiment of the present invention is a method for delivering an anti-target compound to a subject for the treatment of a disorder without being inactivated by administering thereto a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another embodiment of the present invention is a method as described above wherein said target is located in the gut system, and said a polypeptide construct is delivered orally.
  • Another embodiment of the present invention is a method as described above wherein said target is located in vagina! and/or rectal tract, and said a polypeptide construct is delivered to the vaginal and/or rectal tract.
  • Another embodiment of the present invention is a method as described above wherein said target is located in nose, upper respiratory tract and/or lung, and said a polypeptide construct is delivered to nose, upper respiratory tract and/or lung.
  • Another embodiment of the present invention is a method as described above wherein said target is located in intestinal mucosa, and said a polypeptide construct is delivered orally.
  • Another embodiment of the present invention is a method as described above wherein said target is located in the tissues beneath the tongue, and said a polypeptide construct is delivered to the tissues beneath the tongue.
  • Another embodiment of the present invention is a method as described above wherein said target is located in the skin, and said a polypeptide construct is delivered topically.
  • Another embodiment of the present invention is a method as described above wherein said target is in, or accessible via the blood, and said a polypeptide construct is delivered orally, to the vaginal and/or rectal tract, nasally, by inhalation though the mouth or nose, to the tissues beneath the tongue, or topically.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target, for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the gastric environment without being inactivated.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the wall of the intestinal mucosa without being inactivated
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the wall of the nose, upper respiratory tract and/or lung without being inactivated
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the wall of virginal and/or rectal tract without being inactivated
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by a therapeutic compound that is able pass through the tissues beneath the tongue without being inactivated
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by a therapeutic compound that is able pass through the skin without being inactivated
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is TNF-alpha and the disorder is inflammation.
  • Another embodiment of the present invention is a method or polypeptide as described above, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 12 to 14.
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is CEA and the disorder colon cancer.
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is EGFR and the disorder is any of head, neck, lung and colon cancer.
  • Another embodiment of the present invention is a method or polypeptide construct as described above, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 23 to 44
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is antigen of Helicobacter pylori and the disorder is any of indigestion, gastritis.
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is antigen of Mycobacterium tuberculosis and the disorder is tuberculosis.
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is antigen of influenza virus and the disorder is flu.
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is antigen of IgE and the disorder is allergic response.
  • Another embodiment of the present invention is a method or polypeptide construct as described above, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 11
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is antigen of MMP and the disorder is cancer.
  • Another embodiment of the present invention is a method or polypeptide construct as described above, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 15 to 22
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above, wherein said target is antigen of IFN-gamma and the disorder is any of cancer, transplant rejection, auto immune disorder.
  • Another embodiment of the present invention is a method or polypeptide construct as described above, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 45 to 70
  • Another embodiment of the present invention is a method as described above or polypeptide construct as described above wherein said target is any of antigen of Helicobacter pylori, antigen of Mycobacterium tuberculosis, antigen of influenza virus.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor, and at least one single domain antibody directed against a therapeutic target.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor, and at least one therapeutic polypeptide or agent.
  • Another embodiment of the present invention is a polypeptide construct as described above wherein said internalising cellular receptor is Epidermal Growth Factor receptor.
  • Another embodiment of the present invention is a polypeptide as described above wherein a single domain antibody directed against an internalising cellular receptor corresponds to a sequence represented by SEQ ID NO: 23 to 44.
  • Another embodiment of the present invention is a polypeptide construct as described above wherein said internalising cellular receptor is any of LDL receptor, FGF2r, ErbB2r, transferring receptor, PDGr, VEGr, or PsmAr.
  • Another embodiment of the present invention is a polypeptide construct as described above wherein a single domain antibody directed against a therapeutic target, is directed against PDK1.
  • Another embodiment of the present invention is a polypeptide construct as described above use in treating cancer
  • Another embodiment of the present invention is a polypeptide construct as described above wherein a single domain antibody directed against a therapeutic target is directed against any of GSK1, Bad, caspase and Forkhead.
  • Another embodiment of the present invention is a polypeptide construct as described above use in treating cancer.
  • Another embodiment of the present invention is a method for delivering an anti-target therapeutic compound to the interior of a cell comprising administering to a subject a polypeptide construct as described above.
  • Another embodiment of the present invention is a method for delivering an anti-target therapeutic compound to the interior of a cell without being inactivated comprising administering to a subject a polypeptide construct as described above.
  • Another embodiment of the present invention is a method as described above wherein said cell is located in the gut system, and said a polypeptide construct is delivered orally.
  • Another embodiment of the present invention is a method as described above wherein said cell is located in vaginal and/or rectal tract, and said a polypeptide construct is delivered to the vaginal and/or rectal tract.
  • Another embodiment of the present invention is a method as described above wherein said cell is located in nose, upper respiratory tract and/or lung, and said a polypeptide construct is delivered to nose, upper respiratory tract and/or lug.
  • Another embodiment of the present invention is a method as described above wherein said cell is located in intestinal mucosa, and said a polypeptide construct is delivered orally.
  • Another embodiment of the present invention is a method as described above wherein said cell is located in the tissues beneath the tongue, and said a polypeptide construct is delivered to the tissues beneath the tongue.
  • Another embodiment of the present invention is a method as described above wherein said cell is located in the skin, and said a polypeptide construct is delivered topically.
  • Another embodiment of the present invention is a method as described above wherein said cell is in, or accessible via the blood, and said a polypeptide construct is delivered orally, to the vaginal and/or rectal tract, nasally, by inhalation though the mouth or nose, to the tissues beneath the tongue, or topically.
  • Another embodiment of the present invention is a polypeptide construct as described above, or a method as described above, wherein the single domain antibodies are humanized Camelidae VHHs.
  • Another embodiment of the present invention is a polypeptide construct as described above, or a method as described above, wherein said single domain antibody is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length single domain antibody.
  • Another embodiment of the present invention is a polypeptide construct as described above or a method as described above, wherein the polypeptide construct is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length polypeptide construct.
  • Another embodiment of the present invention is a polypeptide construct as described above or a method as described above wherein said single domain antibodies are Camelidae VHHs.
  • Another embodiment of the present invention is a nucleic acid capable of encoding a polypeptide construct as described above.
  • Another embodiment of the present invention is a composition comprising a polypeptide construct as defined above, together with a pharmaceutical carrier.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to a polypeptide construct comprising one or more single domain antibodies directed to one or more target molecule(s), each in a suitable dosage form either directly or as part of a composition containing an ingredient which facilitates delivery.
  • The invention further relates to polypeptide constructs comprising one or more single domain antibodies, for administration to a subject by non-invasive methods, such as orally, sublingually, topically, nasally, vaginally, rectally or by inhalation. Such non-invasive routes of delivery unexpectly provide an effective means to conveniently deliver therapeutic compounds
  • The present invention also relates to constructs comprising one or more single domain antibodies, for administration to a subject by normal invasive methods such as intravenously and subcutaneously.
  • The invention further relates to a method for delivering therapeutic peptides comprises the steps of administering a polypeptide construct comprising one or more single domain antibodies orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation to a subject.
  • The invention further relates to polypeptide constructs comprising anti-IgE single domain antibodies.
  • Single domain antibodies are antibodies whose complementary determining regions are part of a single domain polypetide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies. Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, Ilama, goat, rabbit, bovine. According to one aspect of the invention, a single domain antibody as used herein is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 9404678 for example. For clarity reasons, this variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins. Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, Ilama, dromedary, alpaca and guanaco. Other species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are within the scope of the invention.
  • VHHs, according to the present invention, and as known to the skilled addressee are heavy chain variable domains derived from immunoglobulins naturally devoid of light chains such as those derived from Camelidae as described in WO 94/04678 (and referred to hereinafter as VHH domains or nanobodies). VHH molecules are about 10x smaller than IgG molecules. They are single polypeptides and very stable, resisting extreme pH and temperature conditions. Moreover, they are resistant to the action of proteases which is not the case for conventional antibodies. Furthermore, in vitra expression of VHHs produces high yield, properly folded functional VHHs. In addition, antibodies generated in Camelids will recognize epitopes other than those recognised by antibodies generated in vitro through the use of antibody libraries or via immunisation of mammals other than Camelids ( WO 9749805 ). As such, anti-albumin VHH's may interact in a more efficient way with serum albumin which is known to be a carrier protein. As a carrier protein some of the epitopes of serum albumin may be inaccessible by bound proteins, peptides and small chemical compounds. Since VHH's are known to bind into 'unusual' or non-conventional epitopes such as cavities ( WO 97/49805 ), the affinity of such VHH's to circulating albumin may be increased.
  • The present invention further relates to a polypeptide construct, wherein a single domain antibody is a VHH directed against a target, wherein the VHH belongs to a class having human-like sequences. The class is characterised in that the VHHs carry an amino acid from the group consisting of glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, tyrosine, tryptophan, methionine, serine, threonine, asparagine, or glutamine at position 45, such as, for example, L45 according to the Kabat numbering. A VHH sequence represented by SEQ ID NO: 15 which binds to MMP-12, belongs to this human-like class of VHH polypeptides. As such, peptides belonging to this class show a high amino acid sequence homology to human VH framework regions and said peptides might be administered to a human directly without expectation of an unwanted immune response therefrom, and without the burden of further humanisation.
  • Another human-like class of Camelidae single domain antibodies represented by sequences 68 which binds to if gamma , have been described in WO030356 and contain the hydrophobic FR2 residues typically found in conventional antibodies of human origin or from other species, but compensating this loss in hydrophilicity by the charged arginine residue on position 103 that substitutes the conserved tryptophan residue present in VH from conventional antibodies. As such, peptides belonging to these two classes show a high amino acid sequence homology to human VH framework regions and said peptides might be administered to a human directly without expectation of an unwanted immune response therefrom, and without the burden of further humanisation.
  • Any of the VHHs as used by the invention may be of the traditional class or of the classes of human-like Camelidae antibodies. Said antibodies may be directed against whole target or a fragment thereof, or a fragment of a homologous sequence thereof. These polypeptides include the full length Camelidae antibodies, namely Fc and VHH domains, chimeric versions of heavy chain Camelidae antibodies with a human Fc domain.
  • Targets of the invention are any which are of pharmaceutical interest. Examples are provided here of several targets, and are not intended to limit the invention thereto. Examples of targets include, TNF-alpha, IgE, IFN-gamma, MMP-12, EGFR, CEA, H.pylori, TB, influenza. A single domain antibody directed against a target means a single domain antibody that is capable of binding to said target with an affinity of better than 10-6M.
  • Targets may also be fragments of said targets. Thus a target is also a fragment of said target, capable of eliciting an immune response. A target is also a fragment of said target, capable of binding to a single domain antibody raised against the full length target.
  • A fragment as used herein refers to less than 100% of the sequence (e.g., 99%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10% etc.), but comprising 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or more amino acids. A fragment is of sufficient length such that the interaction of interest is maintained with affinity of 1 x 10-6 M or better.
  • A fragment as used herein also refers to optional insertions, deletions and substitutions of one or more amino acids which do not substantially alter the ability of the target to bind to a single domain antibody raised against the wild-type target. The number of amino acid insertions deletions or substitutions is preferably up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55: 56, 57, 58, 59, 60, 61, 621 631 64, 65, 66, 67, 68, 69 or 70 amino acids.
  • One embodiment of the present invention is a polypeptide construct as disclosed herein, wherein the number of single domain antibodies directed to a target is two or more. Such multivalent polypeptide constructs have the advantage of unusually high functional affinity for the target, displaying much higher than expected inhibitory properties compared to their monovalent counterparts.
  • Multivalent polypeptide constructs have functional affinities that are several orders of magnitude higher than polypeptide constructs which are monovalent. The inventors have found that the functional affinities of these multivalent polypeptide are much higher than those reported in the prior art for bivalent and multivalent antibodies. Surprisingly, the multivalent polypeptide constructs of the present invention linked to each other directly or via a short linker sequence show the high functional affinities expected theoretically with multivalent conventional four-chain antibodies.
  • The inventors have found that such large increased functional activities can be detected preferably with antigens composed of multidomain and multimeric proteins, either in straight binding assays or in functional assays, e.g. animal model of chronic colitis.
  • A multivalent anti-target polypeptide as used herein refers to a polypeptide comprising two or more anti-target polypeptides which have been covalently linked. The anti-target polypeptides may be identical in sequence or may be different in sequence, but are directed against the same target or antigen. Depending on the number of anti-target polypeptides linked, a multivalent anti-target polypeptide may be bivalent (2 anti-target polypeptides), trivalent (3 anti-target polypeptides), tetravalent (4 anti-target polypeptides) or have a higher valency molecules.
  • An example of a multivalent polypeptide construct of the invention, comprising more than one anti-TNF-alpha VHHs is described in Example 7.
  • The single domain antibodies may be joined to form any of the polypeptide constructs disclosed herein comprising more than one single domain antibody using methods known in the art or any future method. They may be joined non-covalently (e.g. using streptavidin/biotin combination, antibody/tag combination) or covalently. They may be fused by chemical crosslinking by reacting amino acid residues with an organic derivatising agent such as described by Blattler et al, Biochemistry 24,1517-1524; EP294703 . Alternatively, the single domain antibody may be fused genetically at the DNA level i.e. a polynucleotide construct formed which encodes the complete polypeptide construct comprising one or more anti-target single domain antibodies. A method for producing bivalent or multivalent VHH polypeptide constructs is disclosed in PCT patent application WO 96/34103 . One way of joining VHH antibodies is via the genetic route by linking a VHH antibody coding sequences either directly or via a peptide linker. For example, the C-terminal end of the VHH antibody may be linked to the N-terminal end of the next single domain antibody.
  • This linking mode can be extended in order to link additional single domain antibodies for the construction and production of tri-, tetra-, etc. functional constructs.
  • According to one aspect of the present invention, the single domain antibodies are linked to each other via a peptide linker sequence. Such linker sequence may be a naturally occurring sequence or a non-naturally occurring sequence. The linker sequence is expected to be non-immunogenic in the subject to which the multivalent anti-target polypeptide is administered. The linker sequence may provide sufficient flexibility to the multivalent anti-target polypeptide, at the same time being resistant to proteolytic degradation. A non-limiting example of a linker sequences is one that can be derived from the hinge region of VHHs described in WO 96/34103 .
  • The polypeptide constructs disclosed herein may be made by the skilled artisan according to methods known in the art or any future method. For example, VHHs may be obtained using methods known in the art such as by immunising a camel and obtaining hybridomas therefrom, or by cloning a library of single domain antibodies using molecular biology techniques known in the art and subsequent selection by using phage display.
  • According to an aspect of the invention a polypeptide construct may be a homologous sequence of a full-length polypeptide construct. According to another aspect of the invention, a polypeptide construct may be a functional portion of a full-length polypeptide construct. According to another aspect of the invention, a polypeptide construct may be a homologous sequence of a full length polypeptide construct. According to another aspect of the invention, a polypeptide construct may be a functional portion of a homologous sequence of a full length polypeptide construct. According to an aspect of the invention a polypeptide construct may comprise a sequence of a polypeptide construct.
  • According to an aspect of the invention a single domain antibody used to form a polypeptide construct may be a complete single domain antibody (e.g. a VHH) or a homologous sequence thereof. According to another aspect of the invention, a single domain antibody used to form the polypeptide construct may be a functional portion of a complete single domain antibody. According to another aspect of the invention, a single domain antibody used to form the polypeptide construct may be a homologous sequence of a complete single domain antibody. According to another aspect of the invention, a single domain antibody used to form the polypeptide construct may be a functional portion of a homologous sequence of a complete single domain antibody.
  • As used herein, a homologous sequence of the present invention may comprise additions, deletions or substitutions of one or more amino acids, which do not substantially alter the functional characteristics of the polypeptides of the invention. The number of amino acid deletions or substitutions is preferably up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1 Ea, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69 or 70 amino acids.
  • A homologous sequence according to the present invention may be a sequence of an anti-target polypeptide modified by the addition, deletion or substitution of amino acids, said modification not substantially altering the functional characteristics compared with the unmodified polypeptide.
  • A homologous sequence of the present invention may be a polypeptide which has been humanised. The humanisation of antibodies of the new class of VHHs would further reduce the possibility of unwanted immunological reaction in a human individual upon administration.
  • A homologous sequence according to the present invention may be a sequence which exists in other Camelidae species such as, for example, camel, Ilama, dromedary, alpaca, guanaco etc.
  • Where homologous sequence indicates sequence identity, it means a sequence which presents a high sequence identity (more than 70%, 75%, 80%, 85%, 90%, 95% or 98% sequence identity) with the parent sequence and is preferably characterised by similar properties of the parent sequence, namely affinity, said identity calculated using known methods.
  • Alternatively, a homologous sequence may also be any amino acid sequence resulting from allowed substitutions at any number of positions of the parent sequence according to the formula below:
    • Ser substituted by Ser, Thr, Gly, and Asn;
    • Arg substituted by one of Arg, His, Gin, Lys, and Glu;
    • Leu substituted by one of Leu, Ile, Phe, Tyr, Met, and Val;
    • Pro substituted by one of Pro, Gly, Ala, and Thr;
    • Thr substituted by one of Thr, Pro, Ser, Ala, Gly, His, and Gin;
    • Ala substituted by one of Ala, Gly, Thr, and Pro;
    • Val substituted by one of Val, Met, Tyr, Phe, lie, and Leu;
    • Gly substituted by one of Gly, Ala, Thr, Pro, and Ser;
    • Ile substituted by one of Ile, Met, Tyr, Phe, Val, and Leu;
    • Phe substituted by one of Phe, Trp, Met, Tyr, Ile, Val, and Leu;
    • Tyr substituted by one of Tyr, Trp, Met, Phe, Ile, Val, and Leu;
    • His substituted by one of His, Glu, Lys, Gln, Thr, and Arg;
    • Gin substituted by one of Gln, Glu, Lys, Asn, His, Thr, and Arg;
    • Asn substituted by one of Asn, Glu, Asp, Gln, and Ser;
    • Lys substituted by one of Lys, Glu, Gln, His, and Arg;
    • Asp substituted by one of Asp, Glu, and Asn;
    • Glu substituted by one of Glu, Asp, Lys, Asn, Gln, His, and Arg;
    • Met substituted by one of Met, Phe, Ile, Val, Leu, and Tyr,
  • A homologous nucleotide sequence according to the present invention may refer to nucleotide sequences of more than 50, 100, 200, 300, 400, 500, 600, 800 or 1000 nucleotides able to hybridize to the reverse-complement of the nucleotide sequence capable of encoding the patent sequence, under stringent hybridisation conditions (such as the ones described by Sambrook et al., Molecular Cloning, Laboratory Manuel, Cold Spring, Harbor Laboratory press, New York).
  • As used herein, a functional portion refers to a sequence of a single domain antibody that is of sufficient size such that the interaction of interest is maintained with affinity of 1 x 10-6 M or better.
  • Alternatively, a functional portion comprises a partial deletion of the complete amino acid sequence and still maintains the binding site(s) and protein domain(s) necessary for the binding of and interaction with its target.
  • As used herein, a functional portion refers to less than 100% of the complete sequence (e.g., 99%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, 1% etc.), but comprising 5 or more amino acids or 15 or more nucleotides.
  • ANT1-lgE SINGLE DOMAIN ANTIBODIES
  • One aspect of the present invention relates to therapeutic compounds which are suitable for alleviating the symptoms, for the treatment and prevention of allergies. Said therapeutic compounds interact with IgE, and modulate the cascade of immunological responses that is responsible for an allergic response.
  • Another aspect of the present invention relates to the use of anti-IgE single domain antibodies (e.g. VHHs)in the preparation of topical ophthalmic compositions for the treatment of an ocular allergic disorder (Example 2). Given the ease of production and the low cost using bacterial or yeast expression systems for VHHs, for example, compared to production of conventional antibodies in mammalian cells, the economics of preparing such compositions using VHHs of the invention are much more favourable then for conventional antibodies.
  • Ocular penetration and consequently ocular efficacy is highly unexpected with conventional antibodies and derived fragments given their large size. The polypeptide constructs of the invention however are expected to be highly efficient given their high potency, stability combined with a low molecular weigth. Therefore, applications for such indications other than topical can be envisaged with polypeptide constructs of the invention.
  • One embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies directed against IgE.
  • Another embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies directed against IgE, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 11. Said sequences are derived from Camelidae VHHs.
  • The present invention also relates to the finding that a polypeptide construct comprising one or more single domain antibodies directed against IgE and further comprising one or more single domain antibodies directed against one or more serum proteins of a subject, surprisingly has significantly prolonged half-life in the circulation of said subject compared with the half-life of the anti-IgE single domain antibody when not part of said construct. Furthermore, such polypeptide constructs were found to exhibit the same favourable properties of VHHs such as high stability remaining intact in mice, extreme pH resistance, high temperature stability and high target affinity.
  • Another embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies directed against IgE further comprising one or more single domain antibodies directed against one or more serum proteins.
  • The serum protein may be any suitable protein found in the serum of subject, or fragment thereof. In one aspect of the invention, the serum protein is serum albumin, serum immunoglobulins, thyroxine-binding protein, transferrin, or fibrinogen. Depending on the intended use such as the required half-life for effective treatment and/or compartimentalisation of the target antigen, the VHH-partner can be directed to one of the above serum proteins.
  • One aspect of the invention, is a polypeptide construct comprising one or more single domain antibodies directed against IgE, further comprising an anti-serum albumin single domain antibody corresponding to a sequence represented by any of SEQ ID NO: 71 to 84.
  • DELIVERY OF POLYPEPTIDE CONSTRUCTS
  • The aspect of the invention relating to the delivery of polypeptide constructs of the invention is not limited to a polypeptide construct comprising anti-IgE single domain antibodies disclosed herein, but, as shown below, is applicable to any target. The polypeptide constructs may comprise single domain antibodies directed against more than one target, optionally with the variations described above.
    One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the gastric environment without being inactivated.
  • As known by persons skilled in the art, once in possession of said polypeptide construct, formulation technology may be applied to release a maximum amount of VHHs in the right location (in the stomach, in the colon, etc.). This method of delivery is important for treating, prevent and/or alleviate the symptoms of disorder whose targets that are located in the gut system.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of a disorder susceptible to modulation by a therapeutic compound that is able pass through the gastric environment without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies specific for antigen related to the disorder.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the prepararion of a medicament for treating, preventing and/or aleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the gastric environment without being inactivated.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the gut system without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound to the vaginal and/or rectal tract.
  • In a non-limiting example, a formulation according to the invention comprises a polypeptide construct as disclosed herein comprising one or more VHHs directed against one or more targets in the form of a gel, cream, suppository, film, or in the form of a sponge or as a vaginal ring that slowly releases the active ingredient over time (such formulations are described in EP 707473 , EP 684814 , US 5629001 ).
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a therapeutic compound to the vaginal and/or rectal tract, by vaginally and/or rectally administering to a subject a polypeptide construct comprising one or more single domain antibodies specific for antigen related to the disorder.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the prepararion of a medicament for treating, preventing and/or aleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound to the vaginal and/or rectal tract without being inactivated.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the vaginal and/or rectal tract without being inactivated, by administering to the vaginal and/or rectal tract of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by administering to the vaginal and/or rectal tract of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target comprising at least one single domain antibody directed against a target, for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound to the nose, upper respiratory tract and/or lung.
  • In a non-limiting example, a formulation according to the invention, comprises a polypeptide construct as disclosed herein directed against one or more targets in the form of a nasal spray (e.g. an aerosol) or inhaler. Since the construct is small, it can reach its target much more effectively than therapeutic IgG molecules.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a therapeutic compound to the upper respiratory tract and lung, by administering to a subject a polypeptide construct as disclosed herein wherein one or more single domain antibodies are specific for an antigen related to the disorder, by inhalation through the mouth or nose.
  • Another aspect of the invention is a dispersible VHH composition, in particular dry powder dispersible VHH compositions, such as those described in US 6514496 . These dry powder compositions comprise a plurality of discrete dry particles with an average particle size in the range of 0.4-10 µm. Such powders are capable of being readily dispersed in an inhalation device. VHH's are particularly suited for such composition as lyophilized material can be readily dissolved (in the lung subsequent to being inhaled) due to its high solubilisation capacity ( Muyldermans, S., Reviews in Molecular Biotechnology, 74, 277-303, (2001)). Alternatively, such lyophilized VHH formulations can be reconstituted with a diluent to generate a stable reconstituted formulation suitable for subcutaneous administration. For example, anti-lgE antibody formulations (Example 1; US 6267958 , EP 841946 ) have been prepared which are useful for treating allergic asthma.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the prepararion of a medicament for treating, preventing and/or aleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound to to the nose, upper respiratory tract and/or lung without being inactivated.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the nose, upper respiratory tract and lung, by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the nose, upper respiratory tract and/or lung without being inactivated, by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders wherein the permeability of the intestinal mucosa is increased. Because of their small size, a polypeptide construct as disclosed herein can pass through the intestinal mucosa and reach the bloodstream more efficiently in subjects suffering from disorders which cause an increase in the permeability of the intestinal mucosa, for example Crohn's disease.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders wherein the permeability of the intestinal mucosa is increased, by orally administering to a subject a polypeptide construct as disclosed herein comprising one or more single domain antibodiesspecific for an antigen related to the disorder.
  • This process can be even further enhanced by an additional aspect of the present invention-the use of active transport carriers. In this aspect of the invention, VHH is fused to a carrier that enhances the transfer through the intestinal wall into the bloodstream. In a non-limiting example, this "carrier" is a second VHH which is fused to the therapeutic VHH. Such fusion constructs made using methods known in the art. The "carrier" VHH binds specifically to a receptor on the intestinal wall which induces an active transfer through the wall.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the prepararion of a medicament for treating, preventing and/or aleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the intestinal mucosa without being inactivated, by administering orally to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by administering orally to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the tissues beneath the tongue effectively. A formulation of said polypeptide construct as disclosed herein, for example, a tablet, spray, drop is placed under the tongue and adsorbed through the mucus membranes into the capillary network under the tongue.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a therapeutic compound that is able pass through the tissues beneath the tongue effectively, by sublingually administering to a subject a VHH specific for an antigen related to the disorder.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the prepararion of a medicament for treating, preventing and/or aleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able to pass through the tissues beneath the tongue.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the tissues beneath the tongue without being inactivated, by administering orally to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inecativated, by administering orally to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the skin effectively. A formulation of said polypeptide construct, for example, a cream, film, spray, drop, patch, is placed on the skin and passes through.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a therapeutic compound that is able pass through the skin effectively, by topically administering to a subject a polypeptide construct as disclosed herein comprising one or more single domain antibodies specific for an antigen related to the disorder.
  • Another aspect of the invention is the use of a polypeptide construct as disclosed herein as a topical ophthalmic composition for the treatment of ocular disorder, such as allergic disorders, which method comprises the topical administration of an ophthalmic composition comprising polypeptide construct as disclosed herein, said construct comprising one or more anti-IgE VHH (Example 1, Example 2).
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the preparation of a medicament for treating, preventing and/or aleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the skin effectively.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the skin without being inactivated, by administering topically to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject, by administering topically to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • A non-limiting example of a therapeutic target against which a polypeptide construct of the invention may be used is TNF, which is involved in inflammatory processes. The blocking of TNF action can have an anti-inflammatory effect, which is highly desirable in certain disease states such as, for example, Crohn's disease. Current therapy consists of intravenous administration of anti-TNF antibodies. Our Examples (Example 4) demonstrate VHH according to the invention which bind TNF and moreover, block its binding to the TNF receptor. Oral delivery of these anti-TNF polypeptide constructs results in the delivery of such molecules in an active form in the colon at sites that are affected by the disorder. These sites are highly inflamed and contain TNF-producing cells. These anti-TNF polypeptide constructs can neutralise the TNF locally, avoiding distribution throughout the whole body and thus limiting negative side-effects. Genetically modified microorganisms such as Micrococcus lactis are able to secrete antibody fragments ( US 6190662 , WO 0023471 ). Such modified microorganisms can be used as vehicles for local production and delivery of antibody fragments in the intestine. By using a strain which produces a TNF neutralizing antibody fragment, inflammatory bowel disorder could be treated. Another aspect of the invention is a polypeptide construct comprising at least one single domain antibody specific for TNF-alpha for use in the treatment, prevention and/or alleviation of disorders relating to inflammatory processes, wherein said polypeptide construct is administered orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to inflammatory processes, comprising administering to a subject a polypeptide construct comprising at least one single domain antibody directed against for example TNF-alpha orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation.
  • According to one aspect of the invention, a polypeptide construct of the invention comprises at least one single domain antibody directed against TNF-alpha, said single domain antibody corresponding to a sequence represented by any of SEQ ID NOs: 12 to 14. Said sequences are anti-TNF-alpha Camelidae VHH.
  • Further non-limiting examples of therapeutic targets against which a polypeptide construct of the invention may be used are certain colon cancer specific antigens, such as, for example, CEA or EGF receptors. In one aspect of the invention, therapeutic VHHs against colon cancer antigens are linked to or provided with one more tumor destroying reagents such as for example, a chemical compound or a radioactive compound.
  • As stated above a colon cancer specific antigen according to the invention is epidermal growth factor receptor (EGFR) which is an essential mediator of cell division in mammalian cells and is a recognised cellular oncogene. After the binding of EGF to its receptor (EGFR), a signaling cascade is initiated resulting in cell development. The EGFR is also involved in human tumorigenesis as it is overexpressed on cells associated with epithelial malignancies located in sites such as the head, neck, lung, colon. Another aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against EGFR for use in the treatment, prevention and/or alleviation of disorders relating to EGFR-mediated cancer, wherein said VHH is administered orally, sublingually, topically, nasally, intravenously, subcutaneously, vaginally, rectally or by inhalation (Examples 25 to 31). Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to EGFR-mediated cancer, comprising administering to a subject a polypeptide construct comprising at least one single domain antibody directed against EGFR orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation.
  • According to one aspect of the invention, a polypeptides construct of the invention comprises at least one single domain antibody directed against EGFR, said single domain antibody corresponding to a sequence represented by any of SEQ ID NOs: 23 to 44. Said sequences are anti-EGRF Camelidae VHHs.
  • As stated above another colon cancer specific antigen according to the invention is carcinoembryonic antigen (CEA), a recognized tumor marker. Another aspect of the invention is a polypeptide construct comprising one or more single domain antibodies specific for CEA for use in the treatment, prevention and/or alleviation of disorders relating to CEA-mediated cancer, wherein said polypeptide is administered orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to CEA-mediated cancer, comprising administering to a subject a polypeptide construct comprising at least one single domain antibody directed against CEA, orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. A few VHHs specific for this glycoprotein have been isolated by selection on solid-phase coated with CEA out of a dedicated library obtained after immunization of a dromedary. By using FACS analysis it appeared that only two fragments recognized the cell-bound antigen. One of the VHHs, that recognised the native structure, has been used to construct a fusion protein with β-lactamase. The functionality of the purified fusion protein was tested in vitro in a prodrug converting cytotoxicity assay. In addition the immunoconjugate was tested in vivo in a tumor-targeting biodistribution study.
  • A non-limiting example of a therapeutic target against which a polypeptide construct of the invention may be used is Helicobacter pylori, which is a bacterium that lives in the mucus which coats the lining of the human stomach and duodenum. The normal human stomach has a very thin layer of mucus that coats the whole of its inside surface. This mucus has a protective role, acting as a barrier between the acid in the stomach and the sensitive stomach wall. H. pylori acts as an irritant to the lining of the stomach, and this causes inflammation of the stomach (gastritis). In one embodiment of the invention is a polypeptide construct comprising at least one single domain antibody directed against H. pylori, said construct and inhibits the enzymatic function of urease. Since single domain antibodies, in particular VHHs have the specific characteristic to occupy enzymatic sites, selected VHHs would inhibit the enzymatic activity and neutralize the virulence of a H, pylori infection. In another aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against H. pylori , said construct inhibiting the adhesion of the bacteria to the stomach wall so preventing irritation of the stomach wall and gastritis. One aspect of the invention is a polypeptide construct comprising one or more single domain antibodies directed against Helicobacter pylori for use in the treatment, prevention and/or alleviation of disorders relating to irritation of the stomach wall and gastritis, wherein said polypeptide construct is administered orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation, but preferably orally. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to irritation of the stomach wall and gastritis, comprising administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against Helicobacter pylori, orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation, but preferably orally.
  • Another non-limiting example of a therapeutic target against which the VHH of the invention may be used is Hepatitis E, which is a viral disorder transmitted via the fecal/oral route. Symptoms increase with age and include abdominal pain, anorexia, dark urine, fever, hepatomegaly, jaundice, malaise, nausea, and vomiting. The overall fatality rate is 1-3%, but 15-25% in pregnant women. Once encountered, most patients develop a neutralizing IgG response which gives life-long protection Neutralizing VHH molecules have the advantage over conventional IgG molecules because they may be administered orally. Since most infections with hepatitis E occur in North-Africa, Central-Africa, Asia and Central-America, oral administration is a significant advantage, since medical logistics are less developed in those countries. One aspect of the invention is one or more VHHs specific for HEV capsid protein (56kDa) for use in the treatment, prevention and/or alleviation of disorders relating hepatitis E, wherein said VHH is administered orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to hepatitis E, comprising administering to a subject said VHH orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation."
    Other non-limiting examples of therapeutic targets against which a polypeptide construct of the invention may be used are micro-organisms induce respiratory disorders such as the TB bacterium and influenza virus. TB or tuberculosis, is a disorder caused by bacteria called Mycobacterium tuberculosis. The bacteria can attack any part of the body, but they usually attack the lungs. Influenza is a viral disorder that causes 'flu'. Influenza viruses are also present in the lung. One aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against Mycobacterium tuberculosis epitope for use in the treatment, prevention and/or alleviation of disorders relating TB, wherein said polypeptide construct is administered orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to TB, comprising administering to a subject said polypeptide construct orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against an influenza virus epitope for use in the treatment, prevention and/or alleviation of disorders relating flu, wherein said polypeptide construct is administered orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to flu, comprising administering to a subject said polypeptide construct orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation.
  • Another non-limiting example of a therapeutic target against which a polypeptide of the invention may be used is IgE in relation to allergies. During their lifetime, subjects may develop an allergic response to harmless parasites (e.g. Dermatophagoides pteronyssinus, house dust mite) or substances (clumps, plastics, metals). This results in the induction of IgE molecules that initiate a cascade of immunological responses. One aspect of the present invention is a polypeptide construct comprising at least one single domain antibody directed against IgE, said polypeptide preventing the interaction of IgE with their receptor(s) on mast cells and basophils. As such they prevent the initiation of the immunological cascade, an allergic reaction. Since IgE molecules are present in the bloodstream, it is within the scope of the invention to fuse the VHH one or more active transport carriers in order to reach their target. Another aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against an IgE epitope for use in the treatment, prevention and/or alleviation of disorders relating to allergies, wherein said polypeptide construct is administered orally, sublingually, topically, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to allergies, comprising administering to a subject said polypeptide construct orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation.
  • According to one aspect of the invention, a polypeptide construct of the invention comprises at least one single domain antibody directed against IgE, said single domain antibody corresponding to a sequence represented by any of SEX ID NOs: 1 to 11. Said sequences are anti-IgE Camelidae VHHs.
  • Another non-limiting example of a therapeutic target against which a polypeptide construct of the invention may be used is human macrophage elastase (MMP-12), which is a member of the family of matrix metalloproteases (MMPs). These enzymes play an important role in normal and inflammatory processes contributing to tissue remodeling and destruction. MMPs play besides proper extracellular matrix remodeling also an important role in diverse disease states such as cancer and inflammation. Macrophage elastase or MMP-12 has a large specificity pocket and broad substrate specificity. It plays a role in several disorders owing to excessive protein degradation of extracellular proteins (e.g. lung damage in smoke induced emphysema, Churg et al, A. 2003) or increased matrix degradation (e.g. higher MMP-12 enzymatic activity in obesity, Chavey et al, 2003). Other clinical indications include coeliac disorder and dermatitis herpetiformis (Salmela et al, 2001), glomerulo nephritis (Kaneko et al, 2003), esophageal squamous cell carcinoma (Ding et al, 2002) and skin cancer (Kerkela et al, 2000).
  • MMP-12 is secreted into the extracellular space by lung alveolar macrophages and dysregulation of MMP-12 is a possible reason for degradation of the alveolar membrane leading to lung emphysema. Target substrates of MMP-12 include extracellular matrix proteins such as elastin, fibronectin and laminin, but also α1-antitrypsin and tissue factor protease inhibitor. One aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against MMP-12 for use in the treatment, prevention and/or alleviation of disorders relating to inflammatory processes, wherein said polypeptide construct is administered orally, sublingually, topically, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to inflammatory processes, comprising administering to a subject said polypeptide construct orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation.
  • Another aspect of this invention consists of (1) VHH's that specifically bind to a metalloproteinase and are not degraded by a metalloproteinase, (2) VHH's which inhibit the proteolytic activity of one or more metalloproteinase and (3) inhibitory VHH's which are highly specific for one MMP (e.g. MMP-12 specific antagonist), unlike none-specific chemical inhibitors ( e.g. batimastat, merimastat...)
  • According to one aspect of the invention, a polypeptide construct of the invention comprises at least one single domain antibody directed against human MMP-12, said single domain antibody corresponding to a sequence represented by any of SEQ ID NOs: 15 to 22. Said sequences are anti-MMP-12 Camelidae VHHs.
  • Another non-limiting example of a therapeutic target against which a polypeptide construct of the invention may be used is IFN-gamma, which is secreted by some T cells. In addition to its anti-viral activity, IFN gamma stimulates natural killer (NK) cells and T helper 1 (Th1) cells, and activates macrophages and stimulates the expression of MHC molecules on the surface of cells. Hence, IFN gamma generally serves to enhance many aspects of immune function, and is a candidate for treatment of disease states where the immune system is over-active (e.g. Crohn's disease), e.g., autoimmune disorders and organ plant rejection. One aspect of the invention is a polypeptide construct comprising at least one single domain antibody directed against IFN-gamma for use in the treatment, prevention and/or alleviation of disorders relating to the immune response, wherein said polypeptide construct is administered orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. Another aspect of the invention is a method of treating, preventing and/or alleviating disorders relating to the immune response, comprising administering to a subject said polypeptide construct orally, sublingually, topically, intravenously, subcutaneously, nasally, vaginally, rectally or by inhalation. In other embodiments of the present invention polypeptide constructs that neutralize IFN gamma are used to treat patients with psoriasis.
  • According to one aspect of the invention, a polypeptide construct of the invention comprises at least one single domain antibody directed against IFN-gamma, said single domain antibody corresponding to a sequence represented by any of SEQ ID NOs: 45 to 70. Said sequences are anti-IFN-gamma Camelidae VHHs.
  • The invention also relates to a method of identifying single domain antibodies (e.g. VHHs) harbouring specific sequences which facilitates the delivery or transport of the anti-target single domain antibodies across human or animal tissues (as described in US 6361938 ), including without limitation GIT epithelial layers, alveolar cells, endothelial of the blood-brain barrier, vascular smooth muscle cells, vascular endothelial cells, renal epithelial cells, M cells of the Peyers Patch, and hepatocytes. Furthermore, delivery systems could be used in conjunction with the VHH's of the invention, comprising nanoparticles, microparticles, liposomes, micelles, cyclodextrines. Only small (<600 daltons) and hydrophobic (Partridge et ai, Adv. Drug Delivery Reviews, 15, 5-36 (1995)) molecules can easily pass the blood-brain barrier, severely limiting the development of novel brain drugs which can be used without the use of invasive neurosurgical procedures.
  • DELIVERING POLYPEPTIDE CONSTRUCTS TO THE INTERIOR OF CELLS
  • Another aspect of the present invention is a method and molecules for delivering therapeutic polypeptides and/or agents to the inside of cells. A further aspect of the invention is a method and molecules for delivering antigens to the inside of antigen presenting cells, and thereby eliciting a powerful immune response thereto. A still further aspect of the invention is to provide a method and molecules for delivery of therapeutic polypeptides and/or agents across natural barriers such as the blood-brain barrier, lung-blood barrier.
  • One aspect of the invention is a polypeptide construct comprising one or more single domain antibodies directed against a target and comprising one or more single domain antibodies directed against an internalising cellular receptor, wherein said polypeptide construct internalises upon binding to said receptor.
  • The targets inside cells may affect the functioning of said cell, or binding thereto may lead to a change in the phenotype of the cell itself by itself. This can be for example, cell death, effects on cell cycling or cell growth or interference with intracellular signaling pathways (see, for example, Poul MA et al, J Mol Biol, 2000, 301, 1149-1161).
  • One embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies specific for an internalising cellular receptor, wherein said construct internalises upon binding to said receptor, wherein the polypeptide construct comprises a therapeutic polypeptide or agent which is covalently or non-covalently linked thereto. Said therapeutic polypeptide or agent has one or more targets which acts intracellularly. See, for example, Figure 12. Said therapeutic polypeptides may harbour specific sequences which target the polypeptide to specific compartiments in the cell, comprising vesicles, organelles and other cytoplasmic structures, membrane-bound structures, the nucleus.
  • An internalising receptor according to the invention is a receptor displayed on the surface of a cell which upon binding to a ligand, mediates the internalisation of said ligand into the cytoplasm of the cell. Internalising receptors according to the invention include, but are not limited to, LDL receptors, EGFr, FGF2r, ErbB2r, transferrin receptor, PDGFr, VEGFr, PsmAr or antigen presenting cell internalising receptors.
  • One embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies specific for an internalising cellular receptor as disclosed herein, further comprising an antigen.
  • One embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies specific for an internalising cellular receptor as disclosed herein, wherein said receptor is an internalising receptor on an antigen presenting cell (APC). Preferably the receptor is highly specific for APCs and not present or is present in lower amounts on other cell types.
  • Another embodiment of the invention is a polypeptide construct comprising one or more anti-receptor single domain antibodies and an antigen. Thus by linking an antigen to a VHH directed towards an internalising receptor on an APC, antigen uptake by APC is not determined by the passive interaction between APC and antigen, but by the "active" binding between VHH and said receptor. This not only makes the process more efficient, but also more reproducible and not dependent on the antigen structure which causes great variability in the T-cell activation from antigen to antigen.
  • After internalization, the complex is digested by the APC and pieces of the antigen can be exposed on the surface in association with MHC/HLA and elicit a more powerful immune response.
  • Another embodiment of the present invention is a method for immunising a subject against an antigen comprising administering to a subject in need thereof a polypeptide construct comprising at least one single domain antibody directed against an antigen present on an APC, wherein said single domain antibody further comprises the antigen of interest.
  • One embodiment of the present invention is a polypeptide construct comprising one or more single domain antibodies specific for an internalising cellular receptor as disclosed herein, wherein said receptor is EGFR. In general internalization of receptors occurs upon binding of the agonistic ligand in a proces called sequestration. In order to ensure that extracellular signals are translated into intracellular signals of appropriate magnitude and specificity, the signalling cascades are tightly regulated via the process of sequestration, whereby receptors are physically removed from the cell surface by internalization to a cytosolic compartment (Carman, C.V. and Benovic, J.L. Current Opinion in Neurobiology 1998, 8: 335-344). This implies that only agonistic ligands or antibodies indeed are expected to internalize via such receptors. In terms of therapeutic use it is not a desired effect that the antibody first triggers proliferation of the tumorcells, before it can deliver a toxic payload to the interiour of the cell.
  • Some of internalising receptors are over-expressed on certain cells, such as the epidermal growth factor receptor (EGFR) or ErBb2 receptor on tumor cells. Epidermal growth factor (EGF) is an essential mediator of cell division in mammalian cells and is a recognized cellular oncogene and is therefore an appropriate target for anti-receptor therapy. After the binding of EGF to its receptor (EGFR), a signaling cascade is initiated resulting in cell development. The EGFR is involved in human tumorigenesis as it is overexpressed on cells of many epithelial malignancies such as head, neck, lung, colon. VHH that are internalised upon binding to one of these receptors can be used to deliver molecules inside the cell.
  • One embodiment of the present invention a polypeptide construct comprising one or more single domain antibodies directed against EGFR, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 23 to 44. Surprisingly, one of the single domain antibodies, did not activate the EGFR, despite the fact that it was internalized efficiently. Such types of antibodies are preferred for therapeutic applications, since these can deliver toxic payloads into cells without stimulating its proliferation.
  • Another embodiment of the present invention is a polypeptide construct construct comprising one or more single domain antibodies directed against for EGFR, wherein said anti-EGFR single domain antibody does not activate the EGFR. Said polypeptide construct may be used for the delivery of a therapeutic agents and/or polypeptides into a cell, as mentioned herein, without stimulating the EGFR.
  • Another embodiment of the present is a polypeptide construct construct comprising one or more single domain antibodies directed against for EGFR, wherein said anti-EGFR single domain antibody does not activate the EGFR and corresponds to a sequence represented by SEQ ID NO: 31.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor, wherein said construct internalises upon binding to said receptor, and further comprising one or more single domain antibodies directed against an intracellular target, said single domain antibodies covalently or non-covalently linked. This multispecific polypeptide construct may be used in the treatment, prevention and/or alleviation of disorders, according to the target of the non-receptor specific single domain antibody. This target can be, for example, a kinase such as PDK1. PDK1 is over-expressed in breast tumor cells. It activates Akt by phosphorylating T308 in the activation loop. A number of downstream substrates of Akt play a direct role in promoting cell survival. These include GSK3, Bad, caspase-9 and Forehead.
  • One embodiment of the present invention is a polypeptide construct comprising a single domain antibody directed against an internalising cellular receptor, wherein said construct internalises upon binding to said receptor, and further comprising one or more single domain antibodies directed against any of PDK1, GSK1, Bad, caspase-9 and Forkhead, Another aspect of the invention the use of said construct for treating cancer. Another aspect of the invention is said construct for the preparation of a medicament for treating cancer.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor, wherein said construct internalizes upon binding to said receptor, wherein the construct further comprises a drug or a toxic compound covalently or non-covalently linked thereto. One example of a toxic compound is a compound that is only active intracellularly due to reducing environment (e.g. an enzyme recombinantly modified with additional cysteins resulting in inactive enzyme, but active in reducing environment). Another example of a toxic compound is a one that is specifically toxic only to a particular cell-type. An example of a toxic compound or a drug is a compound activated by a ligand present inside the cell and leading to the phenotype of interest. Other examples include prodrugs, small organic molecules. One aspect of the invention the use of said construct in the treatment of disorder requiring administration of the same. Another aspect of the invention is said construct for the preparation of a medicament for the treatment of disorder requiring administration of the same.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor, wherein said construct internalises upon binding to said receptor, and wherein a filamentous phage expresses said construct on its surface. Said construct may be attached to the tip of the phage. In one aspect of the invention, construct-phage assembly can be used to package and deliver DNA to the cell for use as a gene therapy vector. According to the invention, the phage may carry DNA in additional to that encoding said construct, for use therapeutically. According to the invention, the phage may carry a gene encoding a therapeutic polypeptide controlled by a promoter for the expression of said gene inside the cell. An example of said promoter includes, but is not limited to, the CMV promoter (Kassner et al, Biochem Biophys Res Commun, 1999, 264: 921-928). Phage have distinct advantages over existing gene therapy vectors because they are simple, economical to produce at high titer, have no intrinsic tropism for mammalian cells, and are relatively simple to genetically modify and evolve (Larocca D et al, Curr. Pharm. Biotechnol, 2002: 3: 45-57).
  • Another embodiment of the present invention is a polypeptide construct as disclosed herein, wherein said single domain antibody is a peptide derived from a VHH specific for an internalising cellular receptor. Said VHH peptide may bind their antigen almost only through the peptide. Internalising VHHs may be prepared from a peptide library which is screened for internalising properties. It is an aspect of the invention that these VHH peptides can be added as a tag to therapeutic polypeptides or agents, for intracellular uptake. The VHH peptide, may, for example, be used to transport a therapeutic VHH into a cell. In one embodiment of the invention, the VHH peptide is the CDR3. In another one embodiment of the invention, the VHH peptide is any other CDR.
  • Another embodiment of the present invention is a method of selecting for VHHs specific for an internalising cellular receptor, wherein said VHH internalise upon binding to said receptor, comprising panning receptor-displaying cells with a phage library (naïve or immune) of VHH, and selecting for internalising VHH by recovering the endocytosed phage from within the cell. The invention includes a selection method which uses cell lines that overexpress a receptor or cell lines transfected with a receptor gene to allow the easy selection of phage antibodies binding to the receptor. This avoids the need for protein expression and purification, speeding up significantly the generation of internalizing VHH.
  • Another embodiment of the present invention is a method for delivering a therapeutic polypeptide, agent or antigen for uptake by cellular internalisation by covalently or non-covalently attaching thereto a polypeptide construct comprising at least one single domain antibody specific for an internalising cellular receptor, wherein said construct internalises upon binding to said receptor.
  • The VHHs according to the invention may be used to treat, prevent and/or alleviate symptoms of disorders requiring the administration of the same.
  • Another embodiment of the present invention is a method for delivering a therapeutic polypeptide or agent that interacts with intracellular targets molecules comprising administering to a subject in need thereof one or more VHHs specific for an internalising cellular receptor, wherein said VHH internalise upon binding to said receptor, wherein said VHH is fused to said polypeptide or agent.
  • Another embodiment of the present invention is a method for delivering a therapeutic polypeptide, agent or antigen across a natural barrier by covalently or non-covalently attaching thereto a polypeptide construct comprising at least one single domain antibody directed against an internalizing cellular receptor, wherein said construct internalises upon binding to said receptor. According to the invention, a natural barrier includes, but is not limited to, the blood-brain, lung-blood, gut-blood, vaginal-blood, rectal-blood and nasal-blood barriers.
  • For example, a peptide construct delivered via the upper respiratory tract and lung can be used for transport of therapeutic polypeptides or agents from the lung lumen to the blood. The construct binds specifically to a receptor present on the mucosal surface (bronchial epithelial cells) resulting in transport, via cellular internalisation, of the therapeutic polypeptides or agents specific for bloodstream targets from the lung lumen to the blood. In another example, a therapeutic polypeptide or agent is linked to a polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor present on the intestinal wall into the bloodstream. Said construct induces a transfer through the wall, via cellular internalization, of said therapeutic polypeptide or agent.
  • Another embodiment of the present invention is a VHH specific for an internalising cellular receptor, wherein said VHH internalises upon binding to said receptor, said VHH is covalently or non-covalently attached to a therapeutic polypeptide or agent, and said VHH crosses a natural barrier.
  • Another embodiment of the present invention is a method for delivering a therapeutic polypeptide, agent or antigen for uptake at a local by covalently or non-covalently attaching it to a VHH specific for an internalising cellular receptor, wherein said VHH internalises upon binding to said receptor. A local area, according to the invention, includes, but is not limited to, the brain, lung, gut, vaginal, rectal and nasal areas.
  • One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the gastric environment without being inactivated.
  • As known by persons skilled in the art, once in possession of said polypeptide construct, formulation technology may be applied to release a maximum amount of VHHs in the right location (in the stomach, in the colon, etc.). This method of delivery is important for treating, prevent and/or alleviate the symptoms of disorder whose targets that are located in the gut system.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of a disorder susceptible to modulation by a therapeutic compound that is able pass through the gastric environment without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies specific for antigen related to the disorder.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the prepararion of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the gastric environment without being inactivated.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the gut system without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by orally administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the vaginal and/or rectal tract.
  • in a non-limiting example, a formulation according to the invention comprises a polypeptide construct as disclosed herein comprising one or more VHHs directed against one or more targets in the form of a gel, cream, suppository, film, or in the form of a sponge or as a vaginal ring that slowly releases the active ingredient over time (such formulations are described in EP 707473 , i=P 684814, US 5629001 ).
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the vaginal and/or rectal tract, by vaginally and/or rectally administering to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the vaginal and/or rectal tract without being inactivated.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the vaginal and/or rectal tract without being inactivated, by administering to the vaginal and/or rectal tract of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by administering to the vaginal and/or rectal tract of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target comprising at least one single domain antibody directed against a target, for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the nose, upper respiratory tract and/or lung.
  • In a non-limiting example, a formulation according to the invention, comprises a polypeptide construct as disclosed herein directed against one or more targets in the form of a nasal spray (e.g. an aerosol) or inhaler. Since the construct is small, it can reach its target much more effectively than therapeutic IgG molecules.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic delivered to the nose, upper respiratory tract and lung, by administering to a subject a polypeptide construct as disclosed herein wherein one or more single domain antibodies are specific for an antigen related to the disorder, by inhalation through the mouth or nose.
  • Another aspect of the invention is a dispersible VHH composition, in particular dry powder dispersible VHH compositions, such as those described in US 6514496 . These dry powder compositions comprise a plurality of discrete dry particles with an average particle size in the range of 6.4-1 g mm. Such powders are capable of being readily dispersed in an inhalation device. VHH's are particularly suited for such composition as lyophilized material can be readily dissolved (in the lung subsequent to being inhaled) due to its high solubilisation capacity (Muyldermans, S., Reviews in Molecular Biotechnology, 74, 277-303, (2001)). Alternatively, such lyophilized VHH formulations can be reconstituted with a diluent to generate a stable reconstituted formulation suitable for subcutaneous administration. For example, anti-IgE antibody formulations (Example 1; US 6267958 , EP 541946 ) have been prepared which are usefull for treating allergic asthma.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the nose, upper respiratory tract and/or lung without being inactivated.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the nose, upper respiratory tract and lung, by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the nose, upper respiratory tract and/or lung without being inactivated, by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated by administering to the nose, upper respiratory tract and/or lung of a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • One embodiment of the present invention is a polypeptide construct as disclosed herein for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa. Because of their small size, a polypeptide construct as disclosed herein can pass through the intestinal mucosa and reach the bloodstream more efficiently in subjects suffering from disorders which cause an increase in the permeability of the intestinal mucosa, for example, Crohn's disease.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa, by orally administering to a subject a polypeptide construct as disclosed herein.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound delivered to the intestinal mucosa, wherein said disorder increases the permeability of the intestinal mucosa.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the intestinal mucosa without being inactivated, by administering orally to a subject a polypeptide construct of the invention.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by administering orally to a subject a polypeptide construct of the invention.
  • This process can be even further enhanced by an additional aspect of the present invention - the use of active transport carriers. In this aspect of the invention, a polypeptide construct as described herein is fused to a carrier that enhances the transfer through the intestinal wall into the bloodstream. In a non-limiting example, this "carrier" is a VHH which is fused to said polypeptide. Such fusion constructs made using methods known in the art. The "carrier" VHH binds specifically to a receptor on the intestinal wall which induces an active transfer through the wall.
  • One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the tissues beneath the tongue effectively. A formulation of said polypeptide construct as disclosed herein, for example, a tablet, spray, drop is placed under the tongue and adsorbed through the mucus membranes into the capillary network under the tongue.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a therapeutic compound that is able pass through the tissues beneath the tongue effectively, by sublingually, administering to a subject a VHH specific for an antigen related to the disorder.
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able to pass through the tissues beneath the tongue.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the tissues beneath the tongue without being inactivated, by administering orally to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject without being inactivated, by administering orally to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • One embodiment of the present invention is a polypeptide construct comprising at least one single domain antibody for use in treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the skin effectively, A formulation of said polypeptide construct, for example, a cream, film, spray, drop, patch, is placed on the skin and passes through.
  • An aspect of the invention is a method for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by a therapeutic compound that is able pass through the skin effectively, by topically administering to a subject a polypeptide construct as disclosed herein comprising one or more single domain antibodies specific for an antigen related to the disorder.
  • Another aspect of the invention is the use of a polypeptide construct as disclosed herein as a topical ophthalmic composition for the treatment of ocular disorder, such as allergic disorders, which method comprises the topical administration of an ophthalmic composition comprising polypeptide construct as disclosed herein, said construct comprising one or more anti-IgE VHH (Example 1, Example 2).
  • Another embodiment of the present invention is a use of a polypeptide construct as disclosed herein for the preparation of a medicament for treating, preventing and/or alleviating the symptoms of disorders susceptible to modulation by an anti-target therapeutic compound that is able pass through the skin effectively.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the skin without being inactivated, by administering topically to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • An aspect of the invention is a method for delivering an anti-target therapeutic compound to the bloodstream of a subject, by administering topically to a subject a polypeptide construct comprising one or more single domain antibodies directed against said target.
  • Another aspect of the present invention is a method to determine which single domain antibodies (e.g. VHHs) molecules cross a natural barrier into the bloodstream upon administration using, for example, oral, nasal, lung, skin. In a non-limiting example, the method comprises administering a naïve, synthetic or immune single domain antibody phage library to a small animal such as a mouse. At different time points after administration, blood is retrieved to rescue phages that have been actively transferred to the bloodstream. Additionally, after administration, organs can be isolated and bound phages can be stripped off. A non-limiting example of a receptor for active transport from the lung lumen to the bloodstream is the Fc receptor N (FcRn). The method of the invention thus identifies single domain antibodies which are not only actively transported to the blood, but are also able to target specific organs. The method may identify which VHH are transported across the gut and into the blood; across the tongue (or beneath) and into the blood; across the skin and into the blood etc.
  • One aspect of the invention are the single domain antibodies obtained by using said method. According to the invention, said single domain antibody may be used as a single domain antibody in a polypeptide construct of the invention. Said construct, further comprising another single domain antibody, a therapeutic agent, or polypeptide carrier directed against a target accessible via or in the blood may be administered by the route most efficient for said single domain antibody.
  • In general, "therapeutically effective amount", "therapeutically effective dose" and "effective amount" means the amount needed to achieve the desired result or results (such as for instance modulating IFN-gamma binding; treating or preventing inflammation). One of ordinary skills in the art will recognize that the potency and, therefore, an "effective amount can vary for the various compounds that modulate ligand-target binding, such as for instance IFN-gamma binding used in the invention. One skilled in the art can readily assess the potency of the compound.
  • As used herein, the term "compound" refers to a polypeptide construct of the present invention, or a nucleic acid capable of encoding said polypeptide construct.
  • By "pharmaceutically acceptable" is meant a material that is not biologically or otherwise undesirable, i.e., the material may be administered to an individual along with the compound without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the pharmaceutical composition in which it is contained.
  • The polypeptide constructs of the present invention are useful for treating or preventing conditions in a subject and comprises administering a pharmaceutically effective amount of a compound or composition.
  • The polypeptide constructs as disclosed here in are useful for treating or preventing conditions in a subject and comprises administering a pharmaceutically effective amount of a compound combination with another, such as, for example, doxorubicin.
  • The present invention is not limited to the administration of formulations comprising a single compound of the invention. It is within the scope of the invention to provide combination treatments wherein a formulation is administered to a patient in need thereof that comprises more than one compound of the invention.
  • A compound useful in the present invention can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient or a domestic animal in a variety of forms adapted to the chosen route of administration, i.e., parenterally, intravenously, intramuscularly, subcutaneously, to the vaginal and/or rectal tract, nasally, by inhalation though the mouth or nose, to the tissues beneath the tongue, or topically.
  • A compound of the present invention can also be administered using gene therapy methods of delivery. See, e.g., U.S. Patent No. 5,399,346 , which is incorporated by reference in its entirety. Using a gene therapy method of delivery, primary cells transfected with the gene for the compound of the present invention can additionally be transfected with tissue specific promoters to target specific organs, tissue, grafts, tumors, or cells.
  • Thus, the present compound may be administered in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • The tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and devices.
  • The active compound may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form must be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • For topical administration, the present compound may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, hydroxyalkyls or glycols or water-alcohol/glycol blends, in which the present compound can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compound to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392 ), Geria (U.S. Pat. No. 4,992,478 ), Smith et al. (U.S. Pat. No. 4,559,157 ) and Wortzman (U.S. Pat. No. 4,820,508 ).
  • Useful dosages of the compound can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949 .
  • Generally, the concentration of the compound(s) in a liquid composition, such as a lotion, will be from about 0.1-25 wt-%, preferably from about 0.5-10 wt-%. The concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt-%, preferably about 0.5-2.5 wt-%.
  • The amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician. Also the dosage of the compound varies depending on the target cell, tumor, tissue, graft, or organ.
  • The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • An administration regimen could include long-term, daily treatment. By "long-term" is meant at least two weeks and preferably, several weeks, months, or years of duration. Necessary modifications in this dosage range may be determined by one of ordinary skill in the art using only routine experimentation given the teachings herein. See Remington's Pharmaceutical Sciences (Martin, E.W., ed. 4), Mack Publishing Co., Easton, PA. The dosage can also be adjusted by the individual physician in the event of any complication.
  • BRIEF DESCRIPTION OF FIGURES
    • Figure 1 : Schematic illustrating the regions of IgE
    • Figure 2 : ELISA of reference and pepsin-treated TNF3E at pH2.2, pH3.2 and pH4.2 (100% is the signal measured at a 1/100 dilution)
    • Figure 3 : Experimental setting
    • Figure 4 : Capacity of VHH clones to inhibit the proteolytic activity of human catalytic domain of MMP12
    • Figure 5 : ELISA to detect A431 specific antibody titers in llama serum.
    • Figure 6 : Detection of EGFR specific antibody titers in llama serum.
    • Figure 7 : Detection of EGFR specific antibody titers in serum of llama 024 and025 and of llama 026 and 027.
    • Figure 8 : Phage response to EGFR
    • Figure 9 : Amino acid alignment of 31 clones identified by the epitope specific elution selection procedure
    • Figure 10 : Phage ELISA on cells (panel A) or on solid-phase immobilized EGFR (panel B) of the 20 unique EGFR specific clones identified via the epitope specific elution selection procedure
    • Figure 11 : Effect of nanobody EGFR-llla42 on receptor internalization and signalling. Fluorescence microscopy visualization of EGFR-llla42 under conditions that allow internalization, with Her-14 (panel A) or 3T3 (panel B). A Western blot that shows the effect of EGFR-llla42 on receptor tyrosin kinase activity is represented in panel C.
    • Figure 12 : Schematic illustrating a use of VHHs directed towards internalising receptors to deliver therapeutic protein, toxic compound, drug or polynucleotide.
    BRIEF DESCRIPTION OF TABLES
    • Table 1: immunization scheme as described in Example 1
    • Table 2: Presence of insert by PCR with vector specific primers as described in Example 1
    • Table 3: First selection as described in Example 1
    • Table 4: Second selection using the rescued phages from the first selection as described in Example 1
    • Table 5: Second round selection using neutravidine coated tubes as described in Example 1
    • Table 6: Number of clones that score positive for binding to both human lgE and chimeric lgE versus the number of clones tested in ELISA as described in Example 1
    • Table 7: Treatment schedule
    • Table 8: Overview of the libraries, their diversity and % insert derived from different llama's and tissues as described in Example 7 and 8
    • Table 9: Immunization schedule and tissue collections
    • Table 10: Overview of constructed libraries
    • Table 11: Overview of epitope specific elution selection procedure
    • Table 12: Overview of 'internalization' selection procedure
    • Table 13: Primer sequences
    • Table 14: Sequence listing
    EXAMPLES Legend of examples:
  • IgE
    • Example 1: VHH directed against IgE
    • Example 2: Formulation of VHH anti-IgE
    • Example 3: Anti-IgE formulation
  • TNF-alpha
    • Example 4: Selection of anti-TNF-alpha
    • Examples 5: Stability testing of antibody fragments specific for human TNF
    • Examples 6: Oral administration of an anti-human TNFα specific VHH in mice
    • Example 7: Efficacy in an animal model for IBD
  • MMP12
    • Example 8: Immunization
    • Example 9: Repertoire cloning
    • Example 10: Rescue of the library and phage preparation
    • Examples 11: Selection of human MMP-12 specific VHH
    • Example 12: Specificity of selected VHH's
    • Example 13: Diversity of selected VHH's
    • Example 14: Expression and purification of VHH
    • Example 15: Functional characterization of selected VHH's: inhibition of MMP-12 proteolytic activity by a VHH in a colorimetric assay.
    • Example 16: Formulation of anti-MMP12 VHH for pulmomary delivery
  • Interferon gamma
    • Example 17: Immunization
    • Example 18: Repertoire cloning
    • Example 19: Rescue of the library and phage preperation
    • Example 20: Selection of human-IFN gamma VHH
    • Example 21: Diversity of selected VHH's
    • Example 22: Expression and purification of VHH
    • Example 23: Topical applications of anti-IFN gamma VHH's
  • Therapeutic VHH-fragments
    • Example 24: Expression of VHH-CDR3 of anti- TNF alpha VHH#3E
  • EGFR
    • Example 25: immunization
    • Example 26: Evaluation of immune response
    • Example 27: Cloning of the heavy-chain antibody fragment (VHH) repertoire
    • Example 28: Evaluation of the cloned repertoire
    • Example 29: Multiple selection strategies to identify EGFR specific nanobodies
    • Example 30: Characterization of EGFR specific nanobodies
    • Example 31: EGF receptor mediated internalization of nanobodies
  • PDK1
    • Example 32: immunisation of llamas
    • Example 33: Repertoire cloning
    • Example 34: Rescue of the library, phage preparation
    • Example 35: Selection
    • Example 36: Screening
    • Example 37: Screen for internalized VHH
    • Example 38: Screen for VHH inhibiting PDK1-Akt interaction
    • Example 39: Making a bispecific construct
    • Example 40: Endocytosis and lysis of tumor cells
    • Example 41: Calculation of homologies between anti-target-single domain antibodies of the invention
    • Example 42: Construction of a bispecific constructs containing a VHH-CDR3 fragment fused to an anti-serum albumin VHH
    Examples: IgE Example 1: VHH directed against IgE
  • Two Ilama's were immunized with human IgE, Scripps laboratories, Cat nr. 10224. The following immunization schemes were used according to Table 1.
  • Different sources for RNA extraction were used:
    • 150 ml immune blood, between 4 and 10 days after the last antigen injection
    • lymph node biopsy 4 days after the last antigen injection
    Peripheral blood lymphocytes (PBLs) were isolated by centrifugation on a density gradient (Ficoll-Paque Plus Amersham Biosciences). PBLs and lymph node were used to extract total RNA (Chomczynski and Sacchi 1987). cDNA was prepared on 200 µg total RNA with MMLV Reverse Transcriptase (Gibco BRL) using oligo d(T) oligonucleotides (de Haard et al., 1999). The cDNA was purified with a phenol/chloroform extraction, followed by an ethanol precipitation and subsequently used as template to amplify the VHH repertoire.
    In a first PCR, the repertoire of both conventional (1.6 kb) and heavy-chain (1.3 kb) antibody gene segments were amplified using a leader specific primer (5'-GGCTGAGCTCGGTGGTCCTGGCT-3') and the oligo d(T) primer (5'-AACTGGAAGAATTCGCGGCCGCAGGAATTTTTTTTTTTTTTTTTT-3'). The resulting DNA fragments were separated by agarose gel electrophoresis and the 1.3 kb fragment encoding heavy-chain antibody segments was purified from the agarose gel. A second PCR was performed using a mixture of FR1 reverse primers ( WO03/054016 sequences ABL037 to ABL043) and the same oligo d(T) forward primer.
    The PCR products were digested with Sfil (introduced in the FR1 primer) and BstEll (naturally occurring in framework 4). Following gel electrophoresis, the DNA fragments of approximately 400 basepairs were purified from gel and ligated into the corresponding restriction sites of phagemid pAX004 to obtain a library of cloned VHHs after electroporation of Escherichia coli TG1. pAX004 allows the production of phage particles, expressing the individual VHHs as a fusion protein with a c-myc tag, a hexahistidine tag and the genelll product. The percentage insert was determined in PCR using a combination of vector based primers.
    Results are summarized in Table 2.
  • Selections were done using chimaeric IgE instead of human IgE, used for immunization, in order to select for VHH molecules directed against the constant region of IgE. The region interacting with the Fcε-receptor is located in the constant part of IgE, more in particular in the region covered by Cε2-Cε3 as shown in Figure 1.
  • A first selection was performed using the pool of PBL day4, PBL day10 and lymph node day4 libraries for each of the two llama's. Chimaeric IgE was solid phase coated at 5 µg/ml and 0.5 µg/ ml and specific phages were eluted using 0.1 M glycine pH = 2.5.
    The results obtained are shown in Table 3.
  • A second selection was performed using the rescued phages from the first selection using 5 µg/ ml. Chimaeric IgE was solid phase coated at 1 µg/ml and specific phages were eluted using buffy coat cells or lysozyme for 1 hr. Buffy coat cells contain cells expressing the Fcεreceptor, while lysozyme is an irrelevant protein and serves as a control. The results obtained are shown in Table 4.
  • Another second round selection was performed using neutravidine coated tubes and 2 nM biotinylated IgE. Specific phages were eluted using buffy coat cells or iysozyme for 1 hr. Buffy coat cells contain cells expressing the Fcεreceptor, while lysozyme is an irrelevant protein and serves as a control. The results obtained are shown in Table 5.
  • Individual clones obtained from the first round of selection were screened in an ELISA using solid phase coated human IgE or chimaeric IgE. The number of clones that score positive for binding to both human IgE and chimeric IgE versus the number of clones tested in ELISA are summarized in Table 6.
  • Clones were picked which were positive for human and chimaeric IgE binding, amplified by PCR and digested with Hinfl. Hinfl profiles were determined on agarose gel and representative clones for different profiles were sequenced. The sequences obtained are shown in Table 14 SEQ ID NOs: 1 to 11.
  • Example 2: Topical applications of anti-IgE VHH's
  • To obtain anti-allergic pharmaceutical compositions for ophthalmic topical applications, a solution of anti-lgE VHH was prepared as follows:
    • eye drops containing a therapeutic dose of anti-lgE VHH dissolved in 100 mi of sterilized water containing 0.9 g sodium chloride, 0.02 g sodium citrate, 0.02 g methyl parahydroxybenzoate, 0.1 g chlorobutanol and acetic acid suitabe to obtain a pH of 6.5.
    • eye ointment containing a therapeutic dose of anti-lgE VHH was prepared according to the conventional method containing 1.0 g of liquid paraffin and a suitable amount of soft paraffin to obtain a total mixture of 100 g.
    Examples 3: Anti-lgE formulations
  • Anti-lgE VHH's that block binding of IgE to its high-affinity receptor are of potential therapeutic value in the treatment of allergy.
    Highly purified VHH#2H11 was dialysed into formulation buffer, followed by addition of lyoprotectant at an isotonic concentration . Isotonic formulation was performed as follows: VHH#2H11 at 25mglml was formulated in 5mM histidine buffer at pH 6 with 500 moles of sugar per mole antibody. This formulation is reconstututed with BWFI (0.9% benzyl alcohol) at a volume which results in a 100 mg/ml of antibody in 20 mM histidine at pH 6 with an isotonic sugar concentration of 340 nM. The binding activity of the anti-IgE VHH in the isotonic formulations was measured in an lgE receptor inhibition assay. It was found that binding activity was essentially unchanged following storage at 4°C for up to 3 months.
  • TNF-alpha Example 4: Selection of anti-TNF-alpha
  • Two llamas were immunized with 100 µg human TNF-alpha per injection according to the schedule described in Example 1. The libraries (short and long immunization procedure) were constructed and selected with in vitro biotinylated TNF-alpha. The biotinylation was carried out as described by Magni et al (Anal Biochem 2001, 298, 181-188).
  • The incorporation of biotin in TNF was evaluated by SDS-PAGE analysis and detection with Extravidin-alkaline phosphatase conjugate (Sigma).
  • The functionality of the modified protein was evaluated for its ability to bind to the solid phase coated recombinant a p75 receptor. {biotinylation} In the first round of selection 400 ng and 50 ng of biotinylated TNF-alpha was captured on neutravidin (Pierce; 10 µg/ml in PBS) coated on the wells of a microtiter plate (NUNC maxisorb). Phage (1.2×1010 TU-s) were added to the wells and incubated for two hours at room temperature. After washing (20 times with PBS-tween and two times with PBS) bound phage was eluted by adding an excess of receptor (extracellular domain of CD120b or p75; 10 µM) or with cells expressing the intact TNF receptor. Between 30,000 and 100,000 phage clones were eluted with TNF from the library derived from the llama immunized using the rapid scheme, while about 10% of these numbers were obtained when eluted with BSA (3 µM; negative control).
  • From the other library (long immunization scheme) 10-fold high numbers were eluted with receptor and BSA, yielding the same enrichment factor (10) as observed before. New phage was prepared from the elution of 50 ng TNF (rapid immunization scheme) and 400 ng TNF (slow scheme) and used for another round of selection on 400, 50 and 10 ng of captured TNF (input: 1.2×1010 phage per well). Approx. 2.5×107 phage were eluted with receptor (10 µM) from the well containing 400 ng and 50 ng of captured TNF and about 2×106 from the well with 10 ng of TNF, while the negative control (elution with 10 µM of BSA) gave only 5 to 10% of those numbers. The observed numbers of eluted phage suggest that the elution with receptor is specific and that those VHH fragments should be eluted that bind to the receptor binding site of TNF.
  • Individual clones were picked and grown in microtiter plate for the production of VHH in culture supernatants. ELISA screening with TNF captured on Extravidin coated plates revealed about 50% positive clones. HinFl-fingerprint analysis showed that 14 different clones were selected, which were grown and induced on 50 ml scale.
  • Periplasmic fractions were prepared, the VHH fragments purified with IMAC and used in an assay to analyze their antagonistic characteristics, i.e. preventing the interaction of TNF with its receptor. For this purpose the VHH (1 µM and 0.3 µM) was incubated with TNF-alpha (3 and 0.7 nM) for 1.5 hours at room temperature (in 0.2% casein / PBS). 100 µl of this mixture was transferred to a well of a microtiter plate, in which the extracellular domain of the receptor was immobilized. After an incubation of one hour the plate was washed and bound TNF was detected with alkaline phosphatase conjugated streptavidin. Two VHH fragments gave antagonistic profiles similar as obtained with 3 and 0.3 µM intact mAB Remicade (Infliximab; Centercor) in spite of the fact that the VHH is truly monomeric, whereas the dimeric appearance of the mAB probably favors the binding of the trimeric TNF-molecule. Similar experiments showing the efficacy of the VHH were performed using the murine sarcoma cell line WEHI and a human cell line expressing the TNF receptor. The sequences obtained are shown in Table 14 SEQ ID NOs: 12 to 13.
  • Example 5: Stability testing of antibody fragments specific for human TNFα
  • Orally administered proteins are subject to denaturation at the acidic pH of the stomach and as well to degradation by pepsin. We have selected conditions to study the resistance of the VHH TNF3E to pepsin which are supposed to mimick the gastric environment. TNF3E a VHH specific to human TNFα was produced as recombinant protein in E.coli and purified to homogeneity by IMAC and gelfiltration chromatography. The protein concentration after purification was determined spectrophotometrically by using the calculated molar extinction coefficient at 280nm. Diluted solutions at 100 microgram/ml were prepared in Mcllvaine buffer (J. Biol Chem. 49, 1921, 183) at pH 2, pH3 and 4 respectively. These solutions were subsequently incubated for 15 minutes at 37°C, prior the addition of porcine gastric mucosa pepsin at a 1/30 w/w ratio. Sixty minutes after adding the protease a sample was collected and immediately diluted 100-fold in PBS pH7.4 containing 0.1% casein to inactivate the pepsin. Seven additional 3-fold dilutions were prepared from this sample for assessing the presence of functional antibody fragment by ELISA. Identical dilutions prepared from an aliquot collected prior the addition of the protease served as a reference. In the ELISA assay biotinylated TNFα was captured in wells of a microtiter plate coated with neutravidin. For both the pepsin-treated and reference samples similar serial dilutions of the samples were prepared and 100 microliter of those dilutions were added to the wells. After incubation for 1 hour the plates were washed. For the detection of VHH binding to of the captured TNFα a polyclonal rabbit anti-VHH antiserum (R42) and an anti-rabbit IgG alkaline phosphatase conjugate was used. After washing, the plates were developed with para nitrophenyl phosphate. The data plotted in Figure 2 shows similar curves for all of the samples exposed to digestive conditions as well as for the reference samples. This indicates that the VHH 3E essentially retains its functional activity under all of the chosen conditions.
  • Example 6: Oral administration of an anti-human TNFα specific VHH in mice
  • An antibody solution containing the anti-human TNFα specific VHH#TNF3E (100microgram per milliliter in 100-fold diluted PBS) was prepared. Three mice which were first deprived from drinking water for 12 hours and subsequently allowed to freely access the antibody solution during the next two hours. Afterwards the mice were sacrificed and their stomachs were dissected. Immediately the content of the stomachs was collected by flushing the stomach with 500microliter PBS containing 1% BSA. This flushed material was subsequently used to prepare serial three-fold dilutions, starting at a 1/5 dilution from the undiluted material. One hundred microliter of these samples was transferred to individual wells of a microtiter plater coated with human TNFα. After incubation for 1 hour and following extensive washing the presence of immuno-reactive material was assessed with a polyclonal rabbit anti-VHH antiserum (R42) followed by incubation with an anti-rabbit alkaline-phosphatase conjugate.The ELISA was developed with paranitrophenyl phosphate. The ELISA signals obtained after 10 minutes clearly demonstrated the presence of functional VHH TNF3E in the gastric flushings of these mice. By comparing to the standard curve we determined the concentration of the functional antibody fragment in the gastric flushing fluid to be 1.5, 12.6 and 8.6 microgram/ml for the three mice tested.
  • Example 7: Efficacy in an animal model for IBD 1) Animal model of chronic collitis
  • The efficacy of bivalent VHH constructs applied via various routes of administration was assessed in a DSS (dextran sodium sulfate) induced model of chronic colitis in BALB/c mice. This model was originally described by Okayasu et al, [Okayasu et al. Gastroenterology 1990; 98: 694-702] and modified by Kojouharoff et. al, [G. Kojouharoff et al. Clin. Exp. Immunol. 1997; 107: 353-8]. The animals were obtained from Charles River Laboratories, Germany, at an age of 11 weeks and kept in the animal facility until they reached a body weight between 21 and 22 g. Chronic colitis was induced in the animals by four DSS treatment cycles. Each cycle consisted of a DSS treatment interval (7 days) where DSS was provided with the drinking water at a concentration of 5 % (w/v) and a recovery interval (12 days) with no DSS present in the drinking water. The last recovery period was prolonged from 12 to 21 days to provide for an inflammation status rather representing a chronic than an acute inflammation at the time of the treatment. Subsequent to the last recovery interval the mice were randomly assigned to groups of 8 mice and treatment with the VHH-constructs was started. The treatment interval was 2 weeks. One week after the end of the treatment interval the animals were sacrificed, the intestine was dissected and histologically examined. The experimental setting is shown schematically in Figure 3.
  • 2) VHH treatment schedule
  • During the VHH treatment period the mice (8 animals per group) were treated daily for 14 consecutive days with bivalent VHH#3F (VHH#3F-VHH#3F: SEQ ID No. 14) by intra-gastric or intra-venous application of 100 µg bivalent VHH 3F. An additional group of animals was treated rectally with the bivalent VHH#3F every other day for a period of 14 days. In all treatment groups a dose of 100 µg of the bivalent VHH#3F was applied at a concentration of 1 mg/ml in a buffered solution. The negative control groups received 100 µl of PBS under otherwise identical conditions. The treatment schedule is shown in Table 7.
  • 3) Results
  • After the mice were sacrificed the body weight was determined and the colon was dissected. The length of the dissected colon was determined and the histology of the colon was assessed by Haematoxilin-Eosin (HE) stain (standard conditions). As compared to the negative controls (PBS treatment) the groups treated with bivalent nanobody 3F showed a prorogued colon length as well as an improved histological score [G. Kojouharoff et al. Clin. Exp. Immunol. 1997; 107: 353-8] thereby demonstrating efficacy of the treatment.
  • MMP12 Example 8: Immunization
  • One llama's (llama 5) was immunized intramuscularly with recombinant human catalytic domain of MMP12 using an appropriate animal-friendly adjuvant Stimune (Cedi Diagnostics BV, The Netherlands). The recombinant catalytic domain was acquired from Prof. H. Tschesche Universität Bielefeld and was supplied as a 56µg/ml solution in 5mM Tris/HCl pH=7.5, 100mM NaCl, 5mM CaCl2 (Lang, R. et al. (2001). The llama received 6 injections at weekly intervals, the first two injections containing each 10 µg of MMP-12, the last four injections containing each 5 µg of MMP-12. Four days after the last immunization a lymph node biopsy (LN) and a blood samples (PBL1) of 150ml was collected from the animal and serum was prepared. Ten days after the last immunization a second blood sample (PBL2) of 150ml was taken and serum was prepared. Peripheral blood lymphocytes (PBLs), as the genetic source of the llama heavy chain immunoglobulins (HcAbs), were isolated from the blood sample using a Ficoll-Paque gradient (Amersham Biosciences) yielding 5×108 PBLs. The maximal diversity of antibodies is expected to be equal to the number of sampled B-lymphocytes, which is about 10% of the number of PBLs (5×107). The fraction of heavy-chain antibodies in llama is up to 20 % of the number of B-lymphocytes. Therefore, the maximal diversity of HcAbs in the 150 ml blood sample is calculated as 107 different molecules. Total RNA was isolated from PBLs and lymph nodes according to the method of Chomczynski and Sacchi(1987).
  • Examples 9: Repertoire cloning
  • cDNA was prepared on 200 µg total RNA with MMLV Reverse Transcriptase (Gibco BRL) using oligo d(T) oligonucleotides (de Haard et al., 1999). The cDNA was purified with a phenol/chloroform extraction, followed by an ethanol precipitation and subsequently used as template to amplify the VHH repertoire.
    In a first PCR, the repertoire of both conventional (1.6 kb) and heavy-chain (1.3 kb) antibody gene segments were amplified using a leader specific primer (5'-GGCTGAGCTCGGTGGTCCTGGCT-3') and the oligo d(T) primer (5'-AACTGGAAGAATTCGCGGCCGCAGGAATTTTTTTTTTTTTTTTTT-3'). The resulting DNA fragments were separated by agarose gel electrophoresis and the 1.3 kb fragment encoding heavy-chain antibody segments was purified from the agarose gel. A second PCR was performed using a mixture of FR1 reverse primers ( WO03/054016 sequences ABL037 to ABL043) and the same oligo d(T) forward primer.
    The PCR products were digested with Sfil (introduced in the FR1 primer) and BstEll (naturally occurring in framework 4). Following gel electrophoresis, the DNA fragments of approximately 400 basepairs were purified from gel and ligated into the corresponding restriction sites of phagemid pAX004 to obtain a library of cloned VHHs after eiectroporation of Escherichia coli TG1, pAX004 allows the production of phage particles, expressing the individual VHHs as a fusion protein with a c-myc tag, a hexahistidine tag and the genelll product. The diversity obtained after electroporation of TG1 cells is presented in Table 8. The percentage insert was determined in PCR using a combination of vector based primers.
  • Example 10: Rescue of the library and phage preparation
  • The library was grown at 37°C in 10 ml 2xTY medium containing 2% glucose, and 100 µg/ml ampicillin, until the OD600nm reached 0.5. M13K07 phages (1012) were added and the mixture was incubated at 37°C for 2 x 30 minutes, first without shaking, then with shaking at 100 rpm. Cells were centrifuged for 5 minutes at 4,500 rpm at room temperature. The bacterial pellet was resuspended in 50 ml of 2xTY medium containing 100 µg/ml ampicillin and 25 µg/ml kanamycin, and incubated overnight at 37°C with vigorously shaking at 250 rpm. The overnight cultures were centrifuged for 15 minutes at 4,500 rpm at 4°C. Phages were PEG precipitated (20% poly-ethylene-glycol and 1.5 M NaCl) for 30 minutes on ice and centrifuged for 20 minutes at 4,500 rpm. The pellet was resuspended in 1 ml PBS. Phages were again PEG precipitated for 10 minutes on ice and centrifuged for 10 minutes at 14,000 rpm and 4°C. The pellet was dissolved in 1 mi 0.5% skimmed milk or PBS-BSA [1mg/ml](Sigma, Cat Nr A3059).
  • Example 11: Selection of human MMP-12 specific VHH
  • Phages were rescued and prepared as described above in Example 10.
  • Two approaches were followed to obtain MMP-12 specific binders:
  • a. Inactive MMP-12 coated on PVDF Membranes
  • 100ng human MMP-12 catalytic domain (diluted in 33µl PBS) was spotted on small pieces (1cm2) of PVDF (immobilon-P, Millipore, Cat Nr IPVH 15150) following the manufacturers guidelines, resulting in an inactive MMP due to the MeOH fixation. As controls an equal amount of lysozyme (Sigma, Cat Nr L-6876) and 33µl PBS were also spotted and immobilized. The membrane pieces were blocked overnight in 5% skimmed milk at 4°C and were washed 3 times with PBS before the phage preparation was applied (4×109 phages in 1ml [5% skimmed milk]). Phages and membrane pieces (in 1,5ml tubes) were incubated for 3 hrs at room temperature with rotation. Then the membranes were transferred to 15ml tubes and were washed 6 times with 10ml [PBS+0.05%Tween-20].
  • Phages were eluted by exposing the membranes to 500µl TEA [70µl in 5ml H2O] for 10min while rotating. The solution containing the eluted phages was removed and the pH was neutralized with 1M Tris pH=7.5.
    Log phase growing TG1 cells were infected with the eluted phages and serial dilutions were plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies after selection obtained from the MMP-12 coated membrane as compared with the negative control where lysozyme was immobilized. Bacteria from MMP selections showing enrichment were scraped and used for a second round of selection. The bacteria were superinfected with helperphage to produce recombinant phages to do a second selection against MMP-12 (as described in Example 9). MMP-12 was immobilized as above and the membrane was blocked overnight at 4°C in 5% skim milk. Phages (2.5×109 in 1ml) were prepared and exposed to the membranes and further selected for MMP binding as during the first round of selection. Log phase growing TG1 cells were infected with the eluted and pH neutralized phages and plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies from the MMP-12 coated membrane as compared with the negative control (immobilized lysozyme).
  • b. Active MMP-12 coated on Nitrocellulose Membrane
  • 250ng human MMP-12 catalytic domain (Biomol Research laboratories Inc. SE 138-9090) was spotted directly on a piece of Hybond-C extra (Amersham Biosciences, Cat Nr RPN 303E) following the suppliers guidelines. As control an equal volume of PBS was spotted. A 5mm diameter disk, containing the spotted area was cut out from each membrane and was transferred to a 1.5ml tube and blocked overnight at 4°C in 1ml BSA-PBS [1mg/ml]. The disks were washed three times in 15ml PBS and subsequently transferred and exposed to the 200ul phage preparation in a microtiterplate well. The phages were prepared as in Example 9 but were preincubated in BSA-PBS for 15min at room temperature. The disks were washed 5 times with PBS/0.05%Tween-20 and were blocked with PBS-BSA for 2 hrs at room temperature. Phages were eluted by exposing the membranes to 100µl TEA [70µl in 5ml H2O] for 10min while rotating. The solution containing the eluted phages was removed and the pH was neutralized with 1 M Tris pH=7.5,
  • Log phase growing TG1 cells were infected with the eluted phages and plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies after selection on the MMP-12 membrane disk as compared with the negative control (PBS). Bacteria from selections with MMP-12 were scraped and used for a second round of selection.
    The bacteria were superinfected with helperphage to produce recombinant phages to do a second selection against MMP-12 (as described in Example 9). MMP-12 was immobilized as above and the membrane was blocked overnight at 4°C in PBS-BSA [1 mg/ml]. Phages (2.5×109 in 1ml) were prepared and exposed to the membranes and further selected for MMP binding as during the first round of selection. Log phase growing TG1 cells were infected with the eluted and neutralized phages and plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies from the MMP-12 coated membrane as compared with the negative control.
  • Example 12: Specificity of selected VHH's
  • Individual clones were picked, grown in 150 µl 2×TY containing 0.1% glucose and 100 µg/ml ampicillin in a microtiter plate at 37°C until OD600nm= 0.6. Then 1 mM IPTG and 5 mM MgSO4 was added and the culture was incubated 4 hours at 37°C. ELISA was performed on the periplasmic extracts (PE, preparation see Example 13) of the cells to examine specificity of the selected clones.
    To examine the clones selected using solid phase coated human MMP-12, plates were coated with human MMP-12 catalytic domain at a concentration of 1 µg/ml overnight at 4°C. Plates were washed 5 times with PBS/0.05% Tween-20. Wells were blocked with 1 % skimmed milk for 2 hrs at room temperature. Periplasmic extracts (100µl) were applied to the wells and incubated for 1 hour at room temperature. Plates were washed 5 times with PBS/0.05%Tween-20. Detection was performed using anti-c-myc antibody, followed by anti-mouse-HRP and ABTS/H2O2 as substrate. Plates were read at 405nm after 30 minutes incubation at room temperature.
    To examine the clones selected using membrane immobilized human MMP-12, 50ng human MMP-12 catalytic domain samples were spotted on PVDF membrane as described in the manufacturers guidelines. 50ng lysozyme was spotted as a negative control. The membranes were blocked with skimmed milk overnight at 4°C, washed 5 times with PBS and transferred to 1,5ml tubes. Periplasmic extracts (100µl) were tenfold diluted in 1% skimmed milk and 1ml was applied per membrane (2cm2) and rotated for 1 hour at room temperature. Membranes were washed 5 times with PBS/0.05%Tween-20. Detection was performed using anti-c-myc antibody, followed by anti-mouse-HRP and DAP as substrate. Membranes were incubated with substrate at room temperature until clear spots were visible. Seven clones which were found to be MMP-12 specific binders are shown in Table 14 SEQ ID NOs 15 to 21.
    In order to check for non specific binding to other MMPs a similar approach was followed in which 50ng of active catalytic domain of MMP 1, 2, 3, 7, 9 and 13 (all from Biomol Research laboratories Inc) was immobilized on Hybond C-extra. The membranes were blocked with skimmed milk overnight at 4°C, washed 5 times with PBS and transferred to 1,5ml tubes. Periplasmic extracts (100µl) were tenfold diluted in 1% skimmed milk and 1ml was applied per membrane (2cm2) and rotated for 1 hour at room temperature. Membranes were washed 5 times with PBS/0.05%Tween-20. Detection was performed using anti-c-myc antibody, followed by anti-mouse-HRP and DAP as substrate. Membranes were incubated with substrate at room temperature until clear spots were visible. No significant detection of the seven selected VHH clones was observed on any of the MMPs other than MMP-12.
  • Results on binders selected against PVDF membrane immobilized human MMP-12 catalytic domain are presented in Table 14 SEQ ID NOs 15 to 21.
    Results on MMP-12 inhibitors selected via Hybond membrane immobilization are presented in Table 14 SEQ ID NO 22.
  • Example 13: Diversity of selected VHH's
  • PCR was performed using M13 reverse and genlll forward primers. The clones were analyzed using Hinf1 fingerprinting and representative clones were sequenced. Sequence analysis was performed resulting in the sequences which are presented in Table 14 SEQ ID NOs 15 to 21 for lmmobilon-P selections and in Table 14 SEQ ID NO 22 for Hybond-C.
  • Example 14: Expression and purification of VHH
  • Clones were grown in 50 ml 2xTY containing 0.1% glucose and 100 µg/ml ampicillin in a shaking flask at 37°C until OD600nm= 2. 1 mM IPTG and 5 mM MgSO4 was added and the culture was incubated for 3 more hours at 37°C. Cultures were centrifuged for 10 minutes at 4,500 rpm at 4°C. The pellet was frozen overnight at -20°C. Next, the pellet was thawed at room temperature for 40 minutes, re-suspended in 1 ml PBS/1mM EDTA/1M NaCl and shaken on ice for 1 hour. Periplasmic fraction was isolated by centrifugation for 10 minutes at 4°C at 4,500 rpm. The supernatant containing the VHH was loaded on Ni-NTA (Qiagen) and purified to homogeneity on an Äkta FPLC chromatography system (Amersham Biosciences). The VHH were eluted from the Ni-NTA using 25mM citric acid pH=4,0 and directly applied on a cation exchange column equilibrated in 25mM citric acid pH=4,0 (Source 30S in a HR5/5 column, Amersham Biosciences). The VHH were eluted with 1 M NaCl in PBS and further purified on a size exclusion column (Superdex 75 HR10/30, Amersham Biosciences) equilibrated in MMP-12 assay buffer [50mM HEPES, 100mM NaCl, 0,05%Brij-35]. The yield of VHH was calculated according to the extinction coefficient and peak surface area.
  • Example 15: Functional characterization of selected VHH's: inhibition of MMP-12 proteolytic activity by a VHH in a colorimetric assay.
  • VHHs were expressed and purified as described in Example 13. Purified VHH was analyzed for the ability to inhibit human MMP-12 catalytic domain using the MMP-12 Colorimetric Assay Kit for Drug Discovery (AK-402) from BlOMOL Research Laboratories. The experimental method conditions described in the Kit were followed.
    The inhibitor supplied with the Kit (Pl115-9090)was used as positive control at the recommended concentration. VHH were applied at a concentration of 7µM. The assay was performed in the microtiterplate Supplied with the BlOMOL Kit and MMP-12 proteolytic activity was followed in a plate reader (405nm) at 37°C.
    The results of one inhibitory VHH and an inactive VHH are presented in Figure 4 together with a positive control.
    Only one VHH molecule (clone P5-29) from selections using active MMP-12 coated on nitrocellulose (Example 12) showed inhibition of human MMP-12 catalytic domain. All other MMP-12 binders (only clone P5-5 is shown), although they bind MMP-12, did not inhibit MMP-12.
  • Examples 16: Formulation of anti-MMP12 VHH for pulmonary delivery
  • A 100% formulation of antibody was prepared by dissolving 5 mg of VHH in 1.0 ml of deionized water. The pH of the solution was 6.5. A 90% formulation of antibody was prepared by dissolving 4.5 mg of VHH in 1.0 ml of 2mM citrate buffer. A 70% formulation of antibody was prepared by dissolving 3.5 mg of VHH in 1 mg/ml of excipient in 1 ml of citrate buffer at pH 6.5. The various classes of excipients used were as follows: Sugar excipients: sucrose, lactose, mannitol, raffinose and trehalose, Polymeric excipients: ficoll and PVP. Protein excipients:: HSA.
    Dry powders of the above formulations were produced by spray drying using a Buchi Spray Dryer.
    The particle size distribution was measure by centrifugal sedimentation.
  • Interferon-gamma Examples 17: immunization
  • Four llama's (llama 5, 6, 22 and 23) were immunized intramuscularly with human IFN-y (PeproTech Inc, USA, Cat Nr: 300-02) using an appropriate animal-friendly adjuvant Stimune (Cedi Diagnostics BV, The Netherlands). Two llama's (llama 29 and 31) were immunized intramuscularly with mouse IFN-y (Protein Expression & Purification core facility, VIB-RUG; Belgium) using an appropriate animal-friendly adjuvant Stimune (Cedi Diagnostics BV, The Netherlands). The llama's received 6 injections at weekly intervals, the first two injections containing each 100 µg of IFN-y, the last four injections containing each 50 µg of IFN-y. Four days after the last immunization a blood sample (PBL1) of 150ml and a lymph node biopsy (LN) was collected from each animal and sera were prepared. Ten days after the last immunization a second blood sample (PBL2) of 150ml was taken from each animal and sera were prepared. Peripheral blood lymphocytes (PBLs), as the genetic source of the llama heavy chain immunoglobulins (HcAbs), were isolated from the blood sample using a Ficoll-Paque gradient (Amersham Biosciences) yielding 5x108 PBLs. The maximal diversity of antibodies is expected to be equal to the number of sampled B-lymphocytes, which is about 10 % of the number of PBL (5x107). The fraction of heavy-chain antibodies in llama is up to 20 % of the number of 5-lymphocytes. Therefore, the maximal diversity of HcAbs in the 150 ml blood sample is calculate as 107 different molecules. Total RNA was isolated from PBLs and lymph nodes according to the method of Chomczyrsski and Sacchi (1987).
  • Example 18: Repertoire cloning
  • cDNA was prepared on 200 µg total RNA with MMLV Reverse Transcriptase (Gibco BRL) using oligo d(T) oligonucleotides (de Haard et al., 1999). The cDNA was purified with a phenol/chloroform extraction, followed by an ethanol precipitation and subsequently used as template to amplify the VHH repertoire.
  • in a first PCR, the repertoire of both conventional (1.6 kb) and heavy-chain (1.3 kb) antibody gene segments were amplified using a leader specific primer (5'-GGCTGAGCTCGGTGGTCCTGGCT-3') and the oligo d(T) primer (5'-AACTGGAAGAATTCGCGGCCGCAGGAATTTTTTTTTTTTTTTTTT-3'). The resulting DNA fragments were separated by agarose gel electrophoresis and the 1.3 kb fragment encoding heavy-chain antibody segments was purified from the agarose gel. A second PCR was performed using a mixture of FR1 reverse primers ( WO03/054016 sequences ABL037 to ABL043) and the same oligo d(T) forward primer.
    The PCR products were digested with Sfil (introduced in the FR1 primer) and BstEll (naturally occurring in framework 4). Following gel electrophoresis, the DNA fragments of approximately 400 basepairs were purified from gel and ligated into the corresponding restriction sites of phagemid pAX004 to obtain a library of cloned VHHs after electroporation of Escherichia coli TG1, pAX004 allows the production of phage particles, expressing the individual VHHs as a fusion protein with a c-myc tag, a hexahistidine tag and the genelll product. The diversity obtained after electroporation of TG1 cells is presented in Table 1. The percentage insert was determined in PCR using a combination of vector based primers.
  • Example 19: Rescue of the Library and phage preparation
  • The library was grown at 37°C in 10 ml 2xTY medium containing 2% glucose, and 100 µg/ml ampicillin, until the OD600nm reached 0.5. M13K07 phages (1012) were added and the mixture was incubated at 37°C for 2 x 30 minutes, first without shaking, then with shaking at 100 rpm. Cells were centrifuged for 5 minutes at 4,500 rpm at room temperature. The bacterial pellet was resuspended in 50 ml of 2xTY medium containing 100 µg/ml ampicillin and 25 µg/ml kanamycin, and incubated overnight at 37°C with vigorously shaking at 250 rpm. The overnight cultures were centrifuged for 15 minutes at 4,500 rpm at 4°C. Phages were PEG precipitated (20% poly-ethylene-glycol and 1.5 M NaCl) for 30 minutes on ice and centrifuged for 20 minutes at 4,500 rpm. The pellet was resuspended in 1 ml PBS. Phages were again PEG precipitated for 10 minutes on ice and centrifuged for 10 minutes at 14,000 rpm and 4°C. The pellet was dissolved in 1 ml PBS-0.1% casein.
  • Examples 20: Selection of human IFN-y specific VHH
  • Phages were rescued and prepared as described above in example 17
  • Two approaches were followed to obtain IFN-y specific binders:
  • a. Solid phase coated IFN-y
  • Microtiter wells were coated with human IFN-y at different concentrations of 10-0.4 µg/well overnight at 4°C. Plates were washed 5 times with PBS/0.05%Tween-20. Wells were blocked with PBS)+ 1% caseine for 2 hrs at room temperature. Phages were incubated for 2 hrs at room temperature. Wells were washed 20 times with PBS+0.05%Tween-20. The two final washes were performed using PBS. Specific phages were eluted using 1 to 2 µg of IFN-y R1 (R&D Systems, Cat Nr: 673-IR/CF) for 1 hr. As negative control elutions were performed using 10 µg Ovalbumine (Sigma, A2512) as irrelevant protein. Log phase growing TG1 cells were infected with the eluted phages and plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies after selection using the receptor for elution as compared with negative control using ovalbumine for elution. Bacteria from selections showing enrichment were scraped and used for a second round of selection.
    The bacteria were superinfected with helperphage to produce recombinant phages as described in example 3. Microtiter wells were coated with IFN-y at different concentrations of 2-0.1 µg/well overnight at 4°C. Plates were washed 5 times with PBS/0.05%Tween-20. Wells were blocked with PBS+1% caseine for 2 hrs at room temperature. Phages were incubated for 2 hrs at room temperature. Wells were washed 20 times with PBS+0.05%Tween-20. The two final washes were performed using PBS. Specific phages were eluted using 1 to 2 µg of IFN-y R1 or 10 µg Ovalbumine as irrelevant protein for 1 hr, subsequently overnight at 4°C and subsequently, phages were eluted using 0.1 M glycine pH 2.5 for 15 minutes at room temperature and neutralized with 1M Tris-HCl pH=7.5. Log phase growing TG1 cells were infected with the eluted and neutralized phages and plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies after selection using the receptor for elution as compared with negative control using ovalbumine for elution.
  • b. Biotinylated IFN-Y
  • Microtiter wells were coated with neutravidine at a concentration of 2 µg/ml overnight at 4°C. Plates were washed 5 times with PBS/0.05%Tween-20. Wells were blocked with PBS+1% caseine for 2 hrs at room temperature. Biotinylated human IFN-y at a concentration of 100-10 ng/well was captured overnight at 4°C, Plates were washed 5 times with PBS/0.05%Tween-20. Phages were incubated for 2 hrs at room temperature. Wells were washed with PBS+0.05%Tween-20, The two final washes were performed using PBS. Specific phages were eluted using 1 to 2 µg of IFN-y R1 (R&D Systems, Cat Nr: 673-lR/CF) for 1 hr. As negative control elutions were performed using 10 µg Ovalbumine (Sigma, A2512) as irrelevant protein. Log phase growing TG1 cells were infected with the eluted phages and plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies after selection using the receptor for elution as compared with negative control using ovalbumine for elution. Bacteria from selections showing enrichment were scraped and used for a second round of selection. Bacteria were superinfected with helperphage to produce recombinant phages. Microtiter wells were coated with neutravidine at a concentration of 2 µg/ml overnight at 4°C. Plates were washed 5 times with PBS/0.05%Tween-20. Wells were blocked with PBS+1% caseine for 2 hrs at room temperature. Biotinylated human IFN-y at a concentration of 20-2.5 ng/100 µl was captured overnight at 4°C. Plates were washed 5 times with PBSI0.05%Tween-20. Phages were incubated for 2 hrs at room temperature. Wells were washed 20 times with PBS+0.05%Tween-20. The two final washes were performed using PBS. Specific phages were eluted using 1 to 2 µg of IFN-y R1 or 10 µg Ovalbumine as irrelevant protein for 1 hr, subsequently overnight at 4°C and subsequently, phages were eluted using 0.1 M glycine pH 2.5 for 15 minutes at room temperature and neutralized with 1 M Tris-HCl pH=7.5. Log phase growing TG1 cells were infected with the eluted and neutralized phages and plated on selective medium. Enrichment was determined by the number of transfected TG1 colonies after selection using the receptor for elution as compared with negative control using ovalbumine for elution.
  • Example 21: Diversity of selected VHH's
  • PCR was performed using M13 reverse and genlll forward primers. The clones were analyzed using Hint1 fingerprinting and representative clones were sequenced. Sequence analysis was performed resulting in the sequences presented in Table 4 for human IFN-y ( SEQ ID No. 45-70).
  • Examples 22: Expression and purification of VHH
  • Small scale expressions were started after transformation of DNA into WK6 Escherichia coli cells.
    Clones were grown in 50 ml 2xTY containing 0.1 % glucose and 100 µg/ml ampicillin in a shaking flask at 37°C until OD600nm= 2. 1 mM IPTG and 5 mM MgSO4 was added and the culture was incubated for 3 more hours at 37°C. Cultures were centrifuged for 10 minutes at 4,500 rpm at 4°C. The pellet was frozen overnight at -20°C. Next, the pellet was thawed at room temperature for 40 minutes, re-suspended in 1 ml PBS/I mM EDTA/IM NaCl and shaken on ice for 1 hour. Periplasmic fraction was isolated by centrifugation for 10 minutes at 4°C at 4,500 rpm. The supernatant containing the VHH was loaded on TALON (Clontech) and purified to homogeneity. The yield of VHH was calculated according to the extinction coefficient.
  • Examples 23: Topical applications of anti-IFN gamma VHH's
    1. 1: To obtain anti-allergic pharmaceutical compositions for ophthalmic topical applications, a solution of at least one anti-IFN gamma VHH was prepared as follows:
      • eye drops containing a therapeutic dose of anti-iFN gamma VHH dissolved in 100 ml of sterilized water containing 0.9 g sodium chloride, 0.02 g sodium citrate. 0.02 g methyl parahydroxybenzoate, 0.1 g chlorobutanol and acetic acid suitabe to obtain a pH of 6.5.
      • eye ointment containing a therapeutic dose of anti-IFN gamma VHH was prepared according to the conventional method containing 1.0 g of liquid paraffin and a suitable amount of soft paraffin to obtain a total mixture of 100 g.
    2. 2: To obtain anti-inflammatory pharmaceutical applications, topical preparations of the present invention contained at least one anti-IFN gamma VHH and a pharmaceutically acceptable carrier. They were prepared as follows:
      • Preparation of Base Cream
    The reagents for preparing the base cream are as follows (contents for 100 kg base cream): Dimethyl silicon oil (17 kg), Liquid paraffin (9 kg), Stearic acid (7.5 kg), Cetyl alcohol (1 kg), Stearyl alcohol (3 kg), Glycerol (20 kg), Ethylparaben (0.1 kg), Peregal A-20 (0.45 kg), Softener SG (0.85 kg), 0.01 M Phosphate Buffer (pH 7.2)(41.1 kg)
    The stainless steel tank was placed into a thermostat water bath and heated to 80° C., which took approximately 10 minutes. The liquid was thoroughly mixed. Then, emulsifying and homogenizing equipment was placed into the open stainless steel tank, the mixture was stirred for 20 minutes at 3500 rpm until fully emulsified. The temperature of the thermostat water bath was cooled naturally to room temperature, until the mixture became a semi-solid cream. The mixture was being continually stirred. - Preparation of Liquid Antibody Mixture
  • VHH#MP3B1SRA was prepared in accordance with Example 22. The lyophilized antibodies were reconstituted with 0.01 M phosphate buffer (pH 7.2) to a concentration of 2 mg/ml. For 1000 gm of base cream, 45 mg of VHH#MP3B1SRA antibody was added.
  • Therapeutic VHH-fragments Example 24: Expression of a VHH-CDR3 fragment of anti-TNFα VHH#3E
  • The CDR3 region of VHH#3E was amplified by using a sense primer located in the framework 4 region (Forward: CCCCTGGCCCCAGTAGTTATACG) and an anti-sense primer located in the framework 3 region (Reverse: TGTGCAGCAAGAGACGG).
    In order to clone the CDR-3 fragment in pAX10, a second round PCR amplification was performed with following primers introducing the required restriction sites:
    • Reverse primer Sfi1 :
    • GTCCTCGCAACTGCGGCCCAGCCGGCCTGTGCAGCAAGAGACGG
    • Forward primer Not1:
    • GTCCTCGCAACTGCGCGGCCGCCCCCTGGCCCCAGTAGTTATACG
  • The PCR reactions were performed in 50 µl reaction volume using 50pmol of each primer. The reaction conditions for the primary PCR were 11 min at 94 °C, followed by 30/60/120 sec at 94/55/72 °C for 30 cycles, and 5 min at 72°C. All reaction were performed wit 2.5 mM MgCl2 , 200 mM dNTP and 1.25U AmpliTaq God DNA Polymerase (Roche Diagnostics, Brussels, Belgium).
    After cleavage with Sfi1 and Not1 the PCR product was cloned in pAX10.
  • EGFR Example 25: Immunization
  • After approval of the Ethical Committee of the Faculty of Veterinary Medicine (University Ghent, Belgium), 4 llamas (024, 025, 026 and 027) were immunized with the tumor antigen epidermal growth factor receptor (EGFR) according to all current animal welfare regulations. To generate an antibody dependent immune response (Table 9), two animals were injected with intact human vulvar squamous carcinoma cells (A431, ATCC CRL 1555), expressing EGFR on its cell surface, while A431 derived membrane extracts were administered to two other llamas (026 and 027). Each animal received seven doses of subcutaneously administered antigens at weekly intervals (Table 9). When immunizing with intact cells, each dose consisted of 108 freshly harvested A431 cells. The dose for immunization with membrane extracts consisted of vesicles prepared from 108 A431 cells. Vesicles were prepared according to Cohen and colleagues (Cohen S, Ushiro H, Stoscheck C, Chinkers M, 1982. A native 170,000 epidermal growth factor receptor-kinase complex from shed plasma membrane vesicles. J. Biol. Chem. 257:1523-31 Vesicles were stored at -80°C before administration. Two extra injections of eight microgram purified EGFR (Sigma) in an emulsion with the adjuvant Stimune (CEDI Diagnostics B.V., Lelystad, The Netherlands) were administered intramuscularly to llama 025 (Table 9).
  • Example 26: Evaluation of immune response
  • At day 0, 28 and 42, 10 ml of (pre-)immune blood was collected and serum was used to evaluate the induction of the immune responses in the 4 animals. A first ELISA was performed to verify whether the animals generated antibodies that recognized A431 epitopes. After coating a tissue-culture treated 96-well plate with gelatin (0.5% in PBS for 10 minutes), the excess of gelatin was removed and A431 cells were grown overnight in the microwells to confluency. Cells were fixed with 4% paraformaldehyde in PBS for 30 minutes at room temperature. Subsequently, the fixative was blocked with 100mM glycine in PBS for 10 minutes, followed by blocking of the wells with a 4% skim milk-PBS solution, again for 10 minutes. Serum dilutions of immunized animals were applied and A431 specific antibodies were detected with a polyclonal anti-llama antiserum developed in rabbit, followed by a secondary goat anti-rabbit horse radish peroxidase (HRP) conjugate (Dako, Denmark). For all four animals, immunization with intact cells or membrane vesicles resulted in the induction of a significant A431-specific antibody titer (Figure 5).
    To verify whether the induced llama antibodies were EGFR specific, antibody titers in serum was evaluated on mouse fibroblasts expressing human EGFR (Her-14) and compared to the parental mouse fibroblasts cell line NIH3T3 clone 2.2 (3T3), similarly performed as described above (Figure 6). Again, the serum titer of antibodies binding to Her-14 was higher compared to the titer for the parental 3T3 cells, indicating that circulating serum antibodies were EGFR specific.
    Finally, the serum response in immunized animals was verified on solid-phase coated purified EGFR. Purified EGFR (Sigma) and the irrelevant carcino embryonic antigen (CEA, Scripps), both at 1 µg/ml, were immobilized overnight at 4°C in a 96 well Maxisorp plate (Nunc). Wells were blocked with a casein solution (1% in PBS). After addition of serum dilutions, specifically bound immunoglobulins were detected using a rabbit anti-liama antiserum followed by a goat anti-rabbit alkaline phosphatase conjugate (Sigma), showing that for all animals a significant antibody dependent immune response against EGFR was induced (Figure 7).
  • Examples 27: Cloning of the heavy-chain antibody fragment (VHH) repertoire
  • Since little is known on the immunoglobulin ontogeny of camelids, B-cell containing tissues of distinct origin and of different time points were collected for each animal (Table 9). After tissue collection, total RNA was isolated according to the procedure described by Chomczynski and Sacchi. (Chomczynski P and Sacchi N. 1987. Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem 162:156-159). The procedure to clone the VHH repertoire is based on a method described in patent application WO 03/054016 . cDNA was prepared on total RNA with MMLV Reverse Transcriptase (Invitrogen) using oligo d(T) oligonucleotides (de Haard HJ, van Neer N, Reurs A, Hufton SE, Roovers RC, Henderikx P, de Bruine AP, Arends JW, Hoogenboom HR. 1999. A large non-immunized human Fab fragment phage library that permits rapid isolation and kinetic analysis of high affinity antibodies. J. Biol, Chem. 274:18218-30). The amounts of RNA of the distinct tissues used for cDNA synthesis is listed in Table 10. The cDNA was purified with a phenol/chloroform extraction, followed by an ethanol precipitation and subsequently used as template to amplify the VHH repertoire.
    In a first PCR, the repertoire of both conventional (1.6 kb) and heavy chain (1.3 kb) antibody gene segments were amplified using a leader specific primer (ABL002) and ABL010, an oligo d(T) primer (for a list of primers see Table 13). The resulting DNA fragments were separated by agarose gel electrophoresis. The amplified 1.3 kb fragment, encoding heavy chain antibody segments was purified from the agarose gel and used as template in a nested PCR using a mixture of FR1 primers (ABL037-ABL043) and ABL010. The PCR products were digested with Sfil (introduced in the FR1 primer) and BstEll (naturally occurring in FR4). Following gel electrophoresis, the DNA fragment of approximately 400 basepairs was purified from gel and 330 ng of amplified VHH repertoire was ligated into the corresponding restriction sites of one microgram of phagemid pAX004 to obtain a library after electroporation of Escherichia coli TG1. pAX004 allows the production of phage particles, expressing the individual VHHs as a fusion protein with the genelll product. The size of the libraries obtained from the distinct tissues collected from the immunized llamas is described in Table 10. As a quality control, a colony PCR using the M13 reverse and a genelll primer was performed on 24 randomly picked colonies of each library and the percentage of clones containing an insert of the correct size was calculated (Table 10).
  • Examples 28: Evaluation of the cloned repertoire
  • In a polyclonal phage ELISA, the specificity of the cloned phage repertoire was evaluated on EGFR and on an irrelevant antigen (TNFa). To generate recombinant virions expressing the VHH repertoire as fusion proteins with the genelll product, the library was grown at 37°C in 10 ml 2xTY medium containing 2% glucose, and 100 µg/ml ampicillin, until the OD600nm reached 0.5. M13K07 phages (1012) were added and the mixture was incubated at 37°C for 2 x 30 minutes, first without shaking, then with shaking at 100 rpm. Cells were centrifuged for 5 minutes at 4,500 rpm at room temperature. The bacterial pellet was resuspended in 50 ml of 2xTY medium containing 100 µg/ml ampicillin and 25 µg/ml kanamycin, and incubated overnight at 37°C with vigorously shaking at 250 rpm. The overnight cultures were centrifuged for 15 minutes at 4,500 rpm at 4°C and supernatant was used to concentrate the phages. Phages were PEG precipitated (20% poly-ethylene-glycol and 1.5 M NaCI) for 30 minutes on ice and centrifuged for 20 minutes at 4,500 rpm. The pellet was resuspended in 1 ml PBS. Phages were again PEG precipitated for 10 minutes on ice and centrifuged for 10 minutes at 14,000 rpm and 4°C. The pellet was dissolved in 1 ml PBS. One µg/ml of EGFR or TNFα was immobilized in a 96 well Maxisorp plate (Nunc) and incubated overnight at 4°C. Plates were washed 5 times with PBS/0.05%Tween-20 and wells were blocked with a casein solution (1% in PBS) and phage dilutions were added for 2 hrs at room temperature. Bound phages were detected using the anti-M13 gpVlll-HRP conjugated monoclonal antibody (Amersham Biosciences) and ABTS/H2O2 as substrate. Plates were read at 405nm after 15 minutes incubation at room temperature. An example of a phage response from a pool of phages rescued from PBL1 libraries of animals 024 and 025 is depicted in Figure 8.
  • Examples 29: Multiple selection strategies to identify EGFR. specific nanobodies
  • Libraries were rescued by growing the bacteria to logarithmic phase (OD600= 0.5), followed by infection with helper phage to obtain recombinant phages expressing the repertoire of cloned VHHs on tip of the phage as gplll fusion protein (as described in Example 18). When selecting for EGFR specific antibodies, two distinct selection strategies have been followed.
  • Selection by epitope specific elution.
  • A first selection strategy was based on the fact that EGFR can be purified by affinity chromatography through ligand elution. Four different elution conditions, applying an excess of molecules that compete for the ligand binding site or overlapping epitope(s) were carried out (Table 11). When selection was performed on A431 or Her-14 cells, unselected recombinant phages were mixed for 20 minutes at 4°C with 6 x 106 blood cells (mainly monocytes, T- and B-cells) or 2 x 107 3T3s, respectively, to deplete for recombinant phages that recognize common, non EGFR-specific epitopes. Unbound phages were then incubated with EGFR÷ selection cells for 2 hours followed by 6 washes with ice-cold PBS. Phages were subsequently eluted with an excess of EGF ligand, mouse monoclonal 2e9 (Defize LH, Moolenaar WH, van der Saag PT, de Laat SW 1986. Dissociation of cellular responses to epidermal growth factor using anti-receptor monoclonal antibodies. EMBO J. 5:1187-92) or EGFR antagonistic antibodies 225 and 528 (Sato JD, Kawamoto T, Le AD, Mendelsohn J, Polikoff J, Sato GH 1983. Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors. Mol. Biol. Med. 1:511-529). All selection steps were performed at 4°C to avoid receptor mediated phage internalization. Logarithmically grown E. coli TG1 was infected with the eluted phages and grown overnight at 37°C on selective medium 2xTY Ap100 and 2% glucose. Cells were scraped and used in a next round of panning whenever required. Two or three rounds of panning were performed to enrich for EGFR specific recombinant phages (Table 11). Whenever purified antigen was used for selection (Table 11), EGFR was immobilized at 1 µg/ml on Maxisorp microtiter plates.
  • Selection for internalizing VHH fragments
  • A second selection strategy was based on the observation that after binding of the ligand to the receptor, EGFR mediated cell signaling can be downregulated by the mechanism of receptor internalization. To identify recombinant phages that are able to internalize through cell surface molecules, the protocol described by Poul and colleagues (Poul MA, Becerril B, Nielsen UB, Morisson P, Marks JD. 2000. Selection of tumor-specific internalizing human antibodies from phage libraries. J. Moi. Biol. 301:1149-61.) was followed. Unselected recombinant phages were added to approximately 2 X 107 mouse fibroblast 3T3s for 30 minutes at 4°C in ice cold binding medium (bicarbonate buffered DMEM; 10% FCS (featal calf serum); 25mM Hepes), supplemented with 2% skim milk to deplete for non-specific VHHs. Unbound phages were subsequently incubated with pre-cooled EGFR- selection cells (Her-14 or A431) in binding medium for 1.5 hours at 4°C, followed by six washes with ice-cold PBS to remove non-bound phages. Cells were covered with pre-warmed binding medium and immediately transferred to 3T°C for 20 minutes, to allow internalization. Subsequently, cells were cooled down to 4°C and were stripped with mild acid (500mM NaCl; 100mM glycine pH2.5) incubations during 10 minutes to remove surface bound recombinant phages. Cells were released from extracellular matrix by trypsinization. Resuspended cells were then lyzed during 4 minutes with 100mM TEA at 4°C to release internalized phages. Logarithmically grown E. coli TG1 was infected with the eluted phages and grown overnight at 37°C on selective medium (2xTY Ap100 with 2% glucose). The libraries used for a single round of selection on A431 and in parallel on Her-14 are summarized in Table 12.
  • Example 30: Characterization of EGFR specific nanobodies
  • To verify EGFR specificity of individual clones after the epitope specific elution procedure of panning, a phage ELISA was performed on individual clones. 47 randomly picked clones for each selection procedure (1, 2, 3, 4, la and llla; Table 11) were grown to logarithmic phase (OD600= 0.5), followed by infection with helper phage to obtain recombinant phages as described in Example 18. A phage ELISA was performed both on solid-phase coated EGFR (comparing to non-coated well) as on gelatin coated Her-14 cells (comparing to 3T3). The presence of EGFR specific VHH was verified by using approximately 109 recombinant phage particles of each clone before detection with an anti-M13 gpVlll-HRP conjugated monoclonal antibody. With clones that scored positive in phage ELISA on cells and/or on solid-phase immobilized EGFR (Table 11), a Hinfl fingerprint analysis was performed (data not shown). The nucleotide sequence was determined for a representative clone of each distinct fingerprint, resulting in 5, 8, 3, 4, 7, and 4 different sequences for conditions, 1, la, 2, llla, 3 and 4, respectively. Amino acid sequence alignment of these 31 binders (Figure 9) indicated that 20 of them were unique (listed in Table 14 SEQ ID Nos 23 to 42). The EGFR specificity of the 20 unique clones in phage ELISA (both on cells and on solid-phase coated EGFR) is shown in Figure 10.
    For the selection according to the internalization protocol, a phage ELISA on cells with a total of 84 individual clones was performed, similarly as for the clones identified by the epitope specific elution selection procedure. After Hinfl fingerprint analysis, nucleotide sequence determination and amino acid sequence alignment to the above described panel of 20 unique binders (data not shown), 2 new anti-EGFR clones, EGFR-B11 and clone EGFR-F11, were identified (Table 14 SEQ ID NOs: 43 to 44). The EGFR specificity of both clones in phage ELISA on cells is shown in Figure 10, panel A.
  • Example 31: EGF receptor mediated internalization of nanobodies.
  • Her-14 and 3T3 cells were grown overnight on glass cover slips, washed with binding medium (see Example 19) and cooled down to 4 °C for 20 minutes. Phages were prepared of nanobody EGFR-llla42 as described in Example 18 and approximately 1012 recombinant virions, diluted in binding medium supplemented with 2% skim milk, were added to the ice cold cells for 1 hour at 4 °C. Cells were washed once with ice cold PBS to remove non bound phages. Subsequently, the cells were shifted to 37°C for 20 minutes to allow phage internalization and again cooled down to 4°C. Cells were washed twice with PBS. Following, cell surface bound phages were removed by two acid washes with stripping buffer (150 mM NaCl, 125 mM HAc) for seven minutes at room temperature. After two washes with PBS, cells were fixed with 4% paraformaldehyde in PBS for 30 minutes at room temperature, and again washed twice with PBS. Fixed cells were then permeabilized in 0.2% Triton X-100 in PBS for 5 minutes at room temperature, followed by two washes with PBS and remaining fixative was blocked with 100mM glycin in PBS for 10 minutes at room temperature. Cells were washed with PBS.-0.5% (w/v) gelatin and internalized phage was visualized by staining with anti-M13 gpVlll-FITC (Amersham Biosciences) followed by an anti-mouse FITC labeled monoclonal antibody and subsequent visualization by fluorescence microscopy. Figure 11 shows that EGFRllla42 is able to internalize Her-14 (panel A) but not 3T3 cells (panel B). Subsequently, FACS analysis demonstrated that nanobody EGFR-llla42 is able to bind both A431 and Her-1 4, but not 3T3 (data not shown).
    To demonstrate the effect of EGF receptor specific nanobodies on receptor signalling, cells were seeded at 100,000 cells per well in 12-well tissue culture plates in medium (DMEM) containing 10% (v/v) serum. After 8 hours, cells were washed once with medium (MEM) containing low (0.5% v/v) serum and serum-starved overnight in the same medium. The day of the assay, medium was refreshed with binding medium (DMEM/ 0.5% FCS / 25mM Hepes and 2% skim milk) and when appropriate, ligand or nanobody (mono- or bivalent) was added at 37°C. After 15 minutes, cells were quickly cooled down on ice and washed twice with ice-cold PBS (10mM Na-phosphate; 150mM NaCl, pH 7.4). Total cell lysates were prepared by scraping the cells off the plate in 50µl protein sample buffer. Proteins were size-separated on 6% (w/v) poly-acrylamide gels (20 µl loaded per gel on two parallel gels) and blotted to PVDF membrane (Roche). Blots were stained for total amount of EGFR with a rabbit polyclonal antiserum to the receptor (Santa Cruz) and for phosphorylated receptor using a monoclonal anti phospho-tyrosine antibody (PY-20; Transduction Labs), followed by an appropriate in donkey developped and peroxidase conjugated secondary antibody (anti-rabbit or anti-mouse). The detection was performed by enhanced chemoluminiscence using Western LightningTM substrate (Perkin Elmer Life Sciences). Surprisingly, anti-EGFR-llla42 nanobody did not activate EGFR÷ cells deprived from EGFR, indicated by the lack of receptor Tyr kinase phosphorylation (Fig 11, panel C). The positive control, in which EGF was added in two concentrations to the cells, clearly induced phosphorylation of the receptor and thus induces activation of the cells.
  • PDK1 Example 32 (1): immunisation of llamas
  • 2 llamas are immunised with a cocktail of recombinant EGF receptor and with PDK1- The lamas are boosted with a cell line overexpressing the EGF receptor. The immunization schemes are summarised in Table 15.
  • Example 33: Repertoire cloning
  • Different sources for RNA extraction are used:
    • 150 ml immune blood, between 4 and 10 days after the last antigen injection
    • lymph node biopsy 4 days after the last antigen injection
    Peripheral blood lymphocytes (PBLs) are isolated by centrifugation on a density gradient (Ficoll-Paque Plus Amersham Biosciences). PBLs and lymph node are used to extract total RNA (Chomczynski and Sacchi 1987) followed by synthesis of cDNA using a hexanucleotide random primer. The repertoire is amplified using two hinge-specific primers: AACAGTTAAGCTTCCGCTTGCGGCCGCGGAGCTGGGGTGTTCGCTGTGGTGGG and AAGAGTTAAGCTTCCGCTTGCGGCCGGTGGTTGTGGTTTTGGTGTCTTGGGTT and a framework 1 specific primer: GAGGTBCARCTGCAGGASTCYGG. Fragments are digested with Pstl and Notl and cloned into a phagemid vector. The repertoire is transformed in TG1 electrocompetent cells and plated on LB agar plates containing 100 µg/ml ampicillin and 2% glucose. Colonies are screened for the presence of insert by PCR with vector specific primers. Example 34: Rescue of the library, phage preparation
  • Libraries are grown at 37°C in 60 ml 2xTY medium containing 2% glucose, and 100 µg/ml ampicillin, until the OD600nm reached 0.5. M13K07 phages (1012) are added and the mixture is incubated at 37°C for 2 x 30 minutes, first without shaking, then with shaking at 100 rpm. Cells are centrifuged for 10 minutes at 4500 rpm at room temperature. The bacterial pellet is resuspended in 300 ml of 2xTY medium containing 100 µg/ml ampicillin and 25 µg/ml kanamycin, and incubated overnight at 30°C with vigorously shaking at 250 rpm. The overnight cultures are centrifuged for 15 minutes at 10.000 rpm at 4°C. Phages are PEG precipitated (20% poly-ethylene-glycol and 1.5 M NaCl) and centrifuged for 30 minutes at 10.000 rpm. The pellet is resuspended in 20 ml PBS. Phages are again PEG precipitated and centrifuged for 30 minutes at 20,000 rpm and 4°C. The pellet is dissolved in 5 ml PBS.
  • Phages are titrated by infection of TG1 cells at OD600nm= 0.5 and plating on LB agar plates containing 100 µg/ml ampicillin and 2% glucose. The number of transformants indicates the number of phages (pfu). The phages are stored at -80°C with 15% glycerol.
  • Example 35. Selection
  • Immunotubes are coated with 2 µg/ml) EGFR, 2 µg/ml PDK1 or with PBS containing 1% casein. After overnight incubation at 4°C, the tubes are blocked with PBS containing 1% casein, for 3 hours at RT. 200 µl phages of the three libraries of llama 005 and of the three libraries of llama006 are pooled and added to the immunotubes with a final volume of 2 ml in PBS for EGFR and in 50 mM Tris HCl (pH 7.4), 150 mM KCI, 1.0 mM DTT, 1 mM MgCl2 and 0.3 mg/ml BSA for PDK1.
  • After 2 hours incubation at RT, the immunotubes are washed 10x with PBS-Tween and 10x with PBS. Bound phages are eluted with 2 ml 0.2 M glycin buffer pH= 2.4. Eluted phages are allowed to infect exponentially growing TG1 cells, and are then plated on LB agar plates containing 100 µg/ml ampicillin and 2% glucose. Examples of results which might be obtained from the panning are presented in Tables 16 and 17.
  • Examples 36: Screening
  • A microtiter plate is coated with 2 µg/ml EGFR or 2 µg/mf PDK1, overnight at 4°C. Plates are blocked for two hours at room temperature with 300 µl 1% casein in PBS. The plates are washed three times with PBS-Tween. Periplasmic extracts are prepared from single colonies and applied to the wells of the microtiter plate. Plates are washed six times with PBS-Tween, after which binding of VHH is detected by incubation with mouse anti-Histidine mAB 1/1000 in PBS for 1 hour at RT followed by anti-mouse-alkaline phosphatase conjugate 1/2000 in PBS, also for 1 hour at RT. Staining is performed with the substrate PNPP (p-nitrophenyl-phosphate, 2 mg/ml in 1 M diethanolamine, 1mM Mg2SO4, pH9.8) and the signals are measured after 30 minutes at 405 nm. An example of the expected number of positive clones versus the number of clones tested in ELISA for each selection is presented in Table 18.
  • Examples 37: Screen for internalised VHH
  • Individual clones specific for the EGFR are amplified by PCR and cloned in a phage engineered to package the green fluorescent protein reporter gene driven by the CMV promoter (Poul MA et al, J Mol Biol, 1999, 288: 203-211). Phages are prepared and incubated with tumor cells (A431) overexpressing EGFR. Phages that endergo EGFR mediated endocytosis are be measured by GFP expression. 1 VHH (EGFR-21) would be expected to show a very high expression of GFP and would be used for further analysis. In another approach internalised phage is stained with anti-phage antibodies (poly- or monoclonal) after permeabilization of cells by treatment with cold methanol as described by Larocca and colleagues (Larocco et al, Molecular Therapy, 2001, 3: 476 - 484) and by Poul and colleagues (Poul MA et al, J Mol Biol, 1999, 288: 203-211).
  • Examples 38: Screen for VHH inhibiting PDK1-Akt interaction
  • PDK1 is coated in a microtiter plate as described above and after blocking the plates, the wells are incubated with 100 µg/ml Akt for one hour at RT. Then (without washing) 100 µl periplasmic extract is added to those wells and VHH binding is measured as described above. VHH that are not able to bind to PDK1 would be scored as inhibitors for the interaction between PDK1 and Akt. The expected number of inhibiting VHH versus the number of VHH tested in inhibition ELISA is summarized in Table 19.
  • Example 39: Making a bispecific construct
  • A bispecific construct is prepared (Conrath et al, J Bioi Chem, 2001, 276: 7346-7350) of EGFR-21 and 5 different strong inhibiting VHHs (PD-1, PD-7, PD-32, PD-33 and PD-72) for PDK1. Protein is prepared and purified to homogeneity for the 5 bispecific constructs and shown to be stable by western blot analysis.
  • Example 40: Endocytosis and lysis of tumor cells
  • Bispecific constructs are incubated with tumor cells (A431) overexpressing EGFR. All constructs that successfully endocytosed would be shown by confocal microscopy. One of the constructs, EGFR-21-PD-32, would be expected to able to inhibit cell growth and finally lead to cell death.
  • Examples 41: Calculation of homologies between anti-target-single domain antibodies of the invention
  • The degree of amino acid sequence homology between anti-target single domain antibodies of the invention was calculated using the Bioedit Sequence Alignment Editor. The calculations indicate the proportion of identical residues between all of the sequences as they are aligned by ClustalW. (Thompson, J.D., Higgins, D.G. and Gibson, T.J. (1994) CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position specific gap penalties and weight matrix choice. Nucleic Acids Research, submitted, June 1994). Table 20 indicates the fraction homology between anti-TNF-alpha VHHs of the invention. Table 21 indicates the percentage homology between anti-IFN-gamma VHHs of the invention.
  • Example 42: Construction of a bispecific construct containing a VHH-CDR3 fragment fused to an anti-serum albumin VHH
  • A functional portion, the CDR3 region of MP2F6SR, was amplified by using a sense primer located in the framework 4 region (F6 CRD3 Forward:CTGGCCCCAGAAGTCATACC) and an anti-sense primer located in the framework 3 region (F6 CDR3 Reverse primer:TGTGCATGTGCAGCAAACC).
    In order to fuse the CDR-3 fragment with the anti-serum albumin VHH MSA-21, a second round PCR amplification was performed with following primers:
    • F6 CDR3 Reverse primer Sfi1:
    • GTCCTCGCAACTGCGGCCCAGCCGGCCTGTGCATGTGCAGCAAACC
    • F6 CDR3 Forward primer Not1:
    • GTCCTCGCAACTGCGCGGCCGCCTGGCCCCAGAAGTCATACG
  • The PCR reactions were performed in 50 ml reaction volume using 50pmol of each primer, The reaction conditions for the primary PCR were 11 min at 94 °C, followed by 30/60/120 sec at 94/55/72 °C for 30 cycles, and 5 min at 72°C. All reaction were performed wit 2.5 mM MgCl2 , 200 mM dNTP and 1.25U AmpliTaq God DNA Polymerase (Roche Diagnostics, Brussels, Belgium).
  • After cleavage of the VHH gene of MSA clones with restriction enzymes Pstl/BstEll the digested products were cloned in pAX11 to obtain clones with a VHH at the C-terminus of the multicloning site. The clones were examined by PCR using vector based primers. From clones yielding a 650 bp product, DNA was prepared and used as acceptor vector to clone the CDR3 of MP2F6SR, after cleavage of the PCR product with restriction enzymes Sfi1/Not1 to allow N-terminal expression of CDR3 in fusion with a MSA VHH.
  • TABLES
  • Table 1: immunization scheme as described in Example 1
    Day Llama 2 Llama 4
    0 100 µg 100 µg
    7 100 µg
    14 50 µg
    21 50 µg 100 µg
    28 50 µg
    35 50 µg
    42 50 µg
    70 50 µg
    Table 2: Presence of insert by PCR with vector specific primers as described in Example 1
    #days after last injection Source RNA Size of the library % insert
    Llama002 4 Lymph 1.3 x 107 89
    4 PBL 1.9 x 107 95
    10 PBL 1.1 x 109 70
    Llama004 4 PBL 1.7 x 108 96
    4 Lymph 4.9 X 107 95
    10 PBL 2.2 x 106 95
    Table 3: First selection as described in Example 1
    5 µg/ ml 0.5 µg/ ml µg /ml (blanco)
    Llama 2 (pool PBL day4, PBLday10, lymph node day4) 1.4 106 2.7 105 1.5 104
    Enrichment compared to blanco 400 x 18 x
    Llama 4 (pool PBL day4, PBLday10, lymph node day4) 3.3 106 4.5 105 7.2 104
    Enrichment compared to blanco 140 x 6.25 x
    Table 4: Second selection using the rescued phages from the first selection as described in
    1 µg/ ml 1 µg/ ml 0 µg ml 0 µg ml
    Elution buffy coat cells Elution Lysozyme Elution buffy coat cells Elution Lysozyme
    Llama 2 (selection 5 µg/ ml lgE: 400 x enrichment) 1.2 108 1.2 108 6 108 3 103
    Enrichment compared to lysozyme elution No enrichment 2x
    Llama 4 (selection 5 µg/ ml IgE: 140 x enrichment) 1.3 108 2 107 3 103 3 103
    Enrichment compared to lysozyme elution 6.5 x No enrichment
  • Example 1
  • Table 5: Second round selection using neutravidine coated tubes as described in Example 1
    2 nM lgE 2 nM lgE 0 nM lgE 0 nM lgE
    Elution buffy coat cells Elution Lysozyme Elution buffy coat cells Elution Lysozyme
    Llama 2 (selection 5 µg/ ml lgE: 400 x enrichment) 1.5 108 1.5 107 3 105 3 105
    Enrichment compared to lysozyme elution 10 x
    Llama 4 (selection 5 µg/ ml lgE: 140 x enrichment) 3.3 107 2.2 107 3 103 6 103
    Enrichment compared to lysozyme elation 1.5 x
    Table 6: Number of clones that score positive for binding to both human lgE and chimeric lgE versus the number of clones tested in ELISA as described in Example 1
    Selection with 5 µg/ml Selection with 0.5 µg/ml
    Llama 002 39/47 21/47
    Llama 004 45/47 46/47
    Table 7: Treatment schedule
    Group Animals Description Schedule
    1 8 negative control 1 ip daily 100 µl PBS i.p. +
    2 8 negative control 2 rectal every other day 100 µl PBS rectal for 2 weeks
    3 8 negative control 3 intragastric daily 100 µl PBS intragastric for 14 consecutive days
    4 8 positive control 1 dexamethasone 5 µg i.p. for 7 consecutive days
    5 8 positive control 2 IL10 expressing Llactis applied orally once per day for 14 consecutive days
    6 8 bivalent VHH 3F intra-gastric daily 100 µg bivalent VHH 3F2 intragastric on 14 consecutive days
    7 8 bivalent VHH 3F i.p. daily 100 µg bivalent VHH 3F i.p. for 14 consecutive days
    8 8 bivalent VHH 3F rectally 100 µg bivalent VHH 3F rectally in 100 µl PBS every other day for two weeks
    Table 8: Overview of the libraries, their diversity and % insert derived from different lama's and tissues as described in Example 7 and 8
    Animal Antigen Source Titer % Insert
    Llama 5 Human MMP-12 PBL time 1 2.1 108 94%
    Llama 5 Human MMP-12 PBL time 2 7.5 106 92%
    Llama 5 Human MMP-12 Lymph node 7.8 108 100%
    Table 9: Immunization schedule and tissue collections
    Day Llama 024 Llama 025 Llama 026 Llama 027
    0 intact cells intact cells vesicles vesicles
    7 intact cells intact cells vesicles vesicles
    14 intact cells intact cells vesicles vesicles
    21 intact cells intact cells vesicles vesicles
    28 intact cells intact cells vesicles vesicles
    35 intact cells intact cells vesicles vesicles
    42 intact cells intact cells vesicles vesicles
    46 150 ml blood sample (PBL1) 150 ml blood sample (PBL 1) 150 ml blood sample (PBL1) lymph node 150 ml blood sample (PBL1) lymph node
    47 lymph node spleen bone marrow
    49 purified EGFR 150 ml blood sample (PBL2) 150 ml blood sample (PBL2)
    55 purified EGFR
    59 150 ml blood sample (P3L2)
    60 lymph node spleen bone marrow
    Table 10: Overview of constructed libraries
    Animal Tissue RNA(µg) Size (x 108) % insert
    Llama 024 PBL1 200 0.25 83
    Llama 024 Lymph node ileum 40 2.3 78
    Llama 024 Lymph node bow 150 0.17 100
    Llama 024 Bone marrow 97 1.5 83
    Llama 024 Spleen 160 0.16 95
    Llama 025 PBL1 200 0.06 95
    Llama 025 Lymph node (ileum + bow) 200 0.8 96
    Llama 025 Bone marrow 200 0.045 88
    Llama 025 Spleen 200 2 86
    Llama 025 PBL2 200 0.13 83
    Llama 026 PBL1 + lymph node 100 + 200 2.46 85
    Llama 027 PBL1 + lymph node 100 +200 1.08 92
    Table 11: Overview of epitope specific elution selection procedure
    Elution condition Elution molecule Selection : antigen format ΦELISA Her-14 ΦELISA EGFR Binder families
    Round I Round 11 Round III
    1 EGF A431 Her-14 - 1/47 24/47 6
    la EGFR 5/47 23/47 8
    2 2e9 A431 Her-14 - 2/47 32/47 5
    llla EGFR 11/47 32/47 4
    3 225 A431 A431 Her-14 8/47 28/47 5
    EGFR 20/47 31/47
    4 528 A431 A431 Her-14 16/47 10/47 5
    EGFR 22/47 29/47
    Table 12: Overview of 'internalization' selection procedure
    Library Selection cells Selected antibody fragment
    Pool lymph node, bone marrow, spleen and PBL1 (024+025) Her-14 A2
    Pool bone marrow (024+025) A431 A4, A9, 611
    Pool PBL1 (E724+025) A431 F11
    Table 13: Primer sequences
    Name Sequences 5'-3'
    ABL002 GGCTGAGCTCGGTGGTCCTGGCT
    ABL010 AACTGGAAGAATTCGCGGCCGCAGGAATTTTTTTTTTTTTTTTTT
    ABL037
    Figure imgb0001
    ABL038 ABL039
    Figure imgb0002
    Figure imgb0003
    ABL040
    Figure imgb0004
    ABL041
    Figure imgb0005
    ABL042
    Figure imgb0006
    ABL043
    Figure imgb0007
    genelll CCACAGACAGCCCTCATAG
    M13 rev GGATAACAATTTCACACAGG
    Table 14: Sequence listing
    SEQ ID NO NAME SEQUENCE
    Anti-IgE VHH
    1 VHH#2C3
    Figure imgb0008
    2 VHH#4G12
    Figure imgb0009
    3 VHH#2C1
    Figure imgb0010
    4 VHH#2H3
    Figure imgb0011
    5 VHH#2D12
    Figure imgb0012
    6 VEH#2G4
    Figure imgb0013
    7 VHH#4C5
    Figure imgb0014
    8 VHH#4A2
    Figure imgb0015
    9 VHH#2D4
    Figure imgb0016
    10 VHH#2B6
    Figure imgb0017
    11 VHH#2H11
    Figure imgb0018
    Anti-TNF alpha VHH
    12 VHH≠3E-His tag
    Figure imgb0019
    13 VHH≠3F
    Figure imgb0020
    14 VHH#3F/ VHK#3F
    Figure imgb0021
    Human MMP-12 specific VHH
    15 MMP-12 P1-1
    Figure imgb0022
    16 MMP-12 P1-3
    Figure imgb0023
    17 MMP--12 P1-7
    Figure imgb0024
    18 MMP- 12 P1-26
    Figure imgb0025
    19 MMP-12 P1-33
    Figure imgb0026
    20 MMP-12 P1-41
    Figure imgb0027
    21 MMP-12 P1-4
    Figure imgb0028
    22 MMP-12 P5-29
    Figure imgb0029
    Anti-EGFR VHH
    23 EGFR-1.4
    Figure imgb0030
    24 EGFR-1.9
    Figure imgb0031
    25 EGFR-1.33
    Figure imgb0032
    26 EGFR-1.34
    Figure imgb0033
    27 EGFR-1.38
    Figure imgb0034
    28 EGFR-Ia1
    Figure imgb0035
    Figure imgb0036
    29 EGFR-Ia7
    Figure imgb0037
    30 EGFR-Ia15
    Figure imgb0038
    31 EGFR-IIIa42
    Figure imgb0039
    32 EGFR,-2.6
    Figure imgb0040
    33 EGFR-2.20
    Figure imgb0041
    34 EGFR-IIIa5
    Figure imgb0042
    35 EGFR-3.18
    Figure imgb0043
    36 EGFR-3.32
    Figure imgb0044
    37 EGFR-3.34
    Figure imgb0045
    38 EGFR-3.39
    Figure imgb0046
    39 EGFR-3.40
    Figure imgb0047
    40 EGFR-4.11
    Figure imgb0048
    4: EGFR-4.21
    Figure imgb0049
    42 EGFR-4.22
    Figure imgb0050
    43 EGFR-B11
    Figure imgb0051
    44 EGFR-F11
    Figure imgb0052
    Anti - human IFN gamma VHH
    45 MP3D2SRA
    Figure imgb0053
    46 MP3A3SR
    Figure imgb0054
    47 MP3C5SR
    Figure imgb0055
    48 MP3C1SR
    Figure imgb0056
    49 MP3G8SR
    Figure imgb0057
    50 MP3D2BR
    Figure imgb0058
    51 MP3H6SRA
    Figure imgb0059
    52 MP3B4SRA
    Figure imgb0060
    53 MP4E4BR
    Figure imgb0061
    54 MP4H8SR
    Figure imgb0062
    55 MP2F6SR
    Figure imgb0063
    56 MP3D1BR
    Figure imgb0064
    57 MP2B56R
    Figure imgb0065
    58 MP2C1BR
    Figure imgb0066
    59 MP4A12SR
    Figure imgb0067
    60 MP3F4SRA
    Figure imgb0068
    61 MP3D3BR
    Figure imgb0069
    62 MP3E5BR
    Figure imgb0070
    63 MP3C7SRA
    Figure imgb0071
    64 MP2F1BR
    Figure imgb0072
    65 MP2C5BR
    Figure imgb0073
    66 MP2C10BR
    Figure imgb0074
    67 MP2G5SR
    Figure imgb0075
    68 MP3B1SRA
    Figure imgb0076
    69 MP2F10SR
    Figure imgb0077
    70 MP3A7SRA
    Figure imgb0078
    Anti-mouse serum albumin VHH
    71 MSA21
    Figure imgb0079
    72 MSAc16
    Figure imgb0080
    73 MSAc112
    Figure imgb0081
    74 MSAc110
    Figure imgb0082
    75 MSAc214
    Figure imgb0083
    76 MSAc116
    Figure imgb0084
    77 MSAc119
    Figure imgb0085
    78 MSAc15
    Figure imgb0086
    79 MSc111
    Figure imgb0087
    80 MSAc115
    Figure imgb0088
    81 MSAc18
    Figure imgb0089
    82 MSAc17
    Figure imgb0090
    83 MSAc120
    Figure imgb0091
    84 MSAC14
    Figure imgb0092
    Table 15: lmmunisation scheme according to Example 32
    Day of immunization Llama 005 EGFr Llama006 EGFr Liama005 PDK1 Llama006 PDK1
    0 100 µ9 40 µg 40 µg 40 µg
    7 100 µg 40 µg
    14 50 µg 20 µg
    21 50 µg 40 µg 20 µg 40 µg
    28 50 µg 20 µg
    35 50 µg 20 µg
    42 20 µg 20 µg
    70 20 µg 20 µg
    Table 16: Results of panning according to Example 35
    llama Source RNA Elution conditions Pfu EGFr Pfu casein Enrichment
    005 Pool of the 3 0.2 M glycin, pH 1 x 107 1 x 104 1000
    libraries 2.4
    006 Pool of the 3 0.2 M glycin, pH 5 x 106 1 x 104 500
    libraries 2.4
    Table 17: Results of panning according to Example 35
    llama Source RNA Elution conditions Pfu PDK1 Pfu casein Enrichment
    005 Pool of the 3 0.2 M glycin, pH 1 x 108 1 x 104 10000
    libraries 2.4
    006 Pool of the 3 0.2 M glycin, pH 9 x 107 1 x 104 9000
    libraries 2.4
    Table 18: Number of positive clones after screening according to Example 36
    target Llama005 Llama006
    EGFr 26/95 38/95
    PDK1 93/95 87/95
    Table 19: Number of inhibiting VHH vs number of VHH tested in inhibition ELISA according to Example 38.
    target Llama005 Llama006
    PDK1 56/93 63/87
    Figure imgb0093
    Figure imgb0094
  • REFERENCES
  • Okayasu l, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R. A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology 1990;98:694-702.
  • Kojouharoff G, Hans W, Obermeier F, Mannel DN, Andus T, Scholmerich J, Gross V, Falk W. Neutralization of tumour necrosis factor (TNF) but not of IL-1 reduces inflammation in chronic dextran sulphate sodium-induced colitis in mice. Clin Exp Immunol 1997;107:353-8.
  • MMP12
    • Salmela MT, Pender SL, Reunala T, MacDonald T, Saarialho-Kere U. Gut, 2001;48(4):496-502 Parallel expression of macrophage metalloelastase (MMP-12) in duodenal and skin lesions of patients with dermatitis herpetiformis.
    • Chavey C, Mari B, Monthouel MN, Bonnafous S, Anglard P, Van Obberghen E, Tartare-Deckert S. J. Biol. Chem., 2003; 278: 11888 - 11896. Matrix metalloproteinases are differentially expressed in adipose tissue during obesity and modulate adipocyte differentiation.
    • Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J, Shapiro SD, Wright JL. Am. J. Respir. Crit. Care Med., 2003; 167: 1083 - 1089. Macrophage Metalloelastase Mediates Acute Cigarette Smoke-induced Inflammation Via TNF-aipha Release.
    • R Lang, A Kocourek, M Braun, H Tschesche, R Huber, W Bode, K Maskos J Mol Biol, Sep 2001; 312(4): 731-42. Substrate specificity determinants of human macrophage elastase (MMP-12) based on the 1. A crystal structure.
    • Yoshikatsu Kaneko , Minoru Sakatsume, Yuansheng Xie, Takeshi Kuroda, Michiko lgashima Ichiei Narita and Fumitake Gejyo The Journal of Immunology, 2003, 170: 3377-3385. Macrophage Metalloelastase as a Major Factor for Glomerular Injury in Anti-Glomerular Basement Membrane Nephritis
    • Ding Y, Shimada Y, Garrin-Rivas MJ, Itami A, Li Z, Hong T, Maeda M, Komoto I, Kawabe A, Kaganoi J, Imamura M. Oncology 2002;63(4):378-84. Clinicopathological significance of human macrophage metalloelastase expression in esophageal squamous cell carcinoma.
    • Kerkela E, Ala-Aho R, Jeskanen L, Rechardt 0, Grenman R, Shapiro SD, Kahari VM, Saarialho-Kere U. J Invest Dermatol 2000 Jun;114(6):1113-9 Expression of human macrophage metalloelastase (MMP-12) by tumor cells in skin cancer.
    PREFERRED EMBODIMENTS
    1. 1. A polypeptide construct comprising at least one single domain antibody directed against lgE.
    2. 2. A polypeptide construct according to item 1 wherein at least one single domain antibody is a Camelidae VHH.
    3. 3. A polypeptide construct according to items 1 and 2 wherein at least one single domain antibody corresponds to a sequence represented by any of SEQ lD NOs: 1 to 11.
    4. 4. A polypeptide construct according to any of items 1 to 3, wherein the number of anti-lgE single domain antibodies is at least two.
    5. 5. A polypeptide construct according any of items 1 to 4, wherein at least one single domain antibody is a humanized Camelidae VHH..
    6. 6. A polypeptide construct according to any of items 1 to 5, wherein a single domain antibody is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length single domain antibody.
    7. 7. A polypeptide construct according to any of items 1 to 6, wherein the polypeptide construct is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length polypeptide construct.
    8. 8. A nucleic acid encoding a polypeptide construct according to any of items 1 to 7.
    9. 9. A polypeptide construct according to any of items 1 to 7 for treating and/or preventing and/or alleviating disorders relating to inflammatory processes.
    10. 10. Use of a polypeptide construct according to any of items 1 to 7 for the preparation of a medicament for treating and/or preventing and/or alleviating disorders relating to inflammatory reactions.
    11. 11. A method for delivering an anti-target compound to a subject for the treatment of a disorder without being inactivated by administering thereto a polypeptide construct comprising one or more single domain antibodies directed against said target,
    12. 12. A method according to item 11 wherein said target is located in the gut system, and said a polypeptide construct is delivered orally.
    13. 13. A method according to item 11 wherein said target is located in vaginal and/or rectal tract, and said a polypeptide construct is delivered to the vaginal and/or rectal tract.
    14. 14. A method according to item 11 wherein said target is located in nose, upper respiratory tract and/or lung, and said a polypeptide construct is delivered to nose, upper respiratory tract and/or lung.
    15. 15. A method according to item 11 wherein said target is located in intestinal mucosa, and said a polypeptide construct is delivered orally.
    16. 16. A method according to item 11 wherein said target is located in the tissues beneath the tongue, and said a polypeptide construct is delivered to the tissues beneath the tongue.
    17. 17. A method according to item 11 wherein said target is located in the skin, and said a polypeptide construct is delivered topically.
    18. 18. A method according to item 11 wherein said target is in, or accessible via the blood, and said a polypeptide construct is delivered orally, to the vaginal and/or rectal tract, nasally, by inhalation though the mouth or nose, to the tissues beneath the tongue, or topically.
    19. 19. A polypeptide construct comprising at least one single domain antibody directed against a target, for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the gastric environment without being inactivated.
    20. 20. A polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the wall of the intestinal mucosa without being inactivated
    21. 21. A polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the wall of the nose, upper respiratory tract and/or lung without being inactivated
    22. 22. A polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by an anti-target therapeutic compound that is able pass through the wall of virginal and/or rectal tract without being inactivated
    23. 23. A polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by a therapeutic compound that is able pass through the tissues beneath the tongue without being inactivated
    24. 24. A polypeptide construct comprising at least one single domain antibody directed against a target for use in treating, preventing and/or alleviating the symptoms of disorders which are susceptible to modulation by a therapeutic compound that is able pass through the skin without being inactivated
    25. 25. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is TNF-aJpha and the disorder is inflammation.
    26. 26. A method or polypeptide according to item 25, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 12 to 14.
    27. 27. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is CEA and the disorder colon cancer.
    28. 28. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is EGFR and the disorder is any of head, neck, lung and colon cancer.
    29. 29. A method or polypeptide construct according to item 28, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 23 to 44
    30. 30. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is antigen of Helicobacter pylori and the disorder is any of indigestion, gastritis.
    31. 31. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is antigen of Mycobacterium tuberculosis and the disorder is tuberculosis.
    32. 32. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is antigen of influenza virus and the disorder is flu,
    33. 33. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is antigen of lgE and the disorder is allergic response.
    34. 34. A method or polypeptide construct according to item 33, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 1 to 11
    35. 35. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is antigen of MMP and the disorder is cancer.
    36. 36. A method or polypeptide construct according to item 35, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 15 to 22
    37. 37. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24, wherein said target is antigen of IFN-gamma and the disorder is any of cancer, transplant rejection, auto immune disorder.
    38. 38. A method or polypeptide construct according to item 37, wherein a single domain antibody corresponds to a sequence represented by any of SEQ ID NOs: 45 to 70
    39. 39. A method according to any of items 11 to 18 or polypeptide construct according to any of item 19 to 24 wherein said target is any of antigen of Helicobacter pylori, antigen of Mycobacterium tuberculosis, antigen of influenza virus.
    40. 40. A polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor, and at least one single domain antibody directed against a therapeutic target.
    41. 41. A polypeptide construct comprising at least one single domain antibody directed against an internalising cellular receptor, and at least one therapeutic polypeptide or agent.
    42. 42. A polypeptide construct according to items 40 and 41 wherein said internalising cellular receptor is Epidermal Growth Factor receptor.
    43. 43. A polypeptide construct according to item 42 wherein a single domain antibody directed against an internalising cellular receptor corresponds to a sequence represented by SEQ ID NO: 23 to 44.
    44. 44. A polypeptide construct according to items 40 and 41 wherein said internalising cellular receptor is any of LDL receptor, FGF2r, ErbB2r, transferring receptor, PDGr, VEGr, or PsmAr.
    45. 45. A polypeptide construct according to any of items 40 to 44 wherein a single domain antibody directed against a therapeutic target, is directed against PDK1.
    46. 46. A polypeptide construct according to item 45 for use in treating cancer
    47. 47. A polypeptide construct according to any of items 40 to 44 wherein a single domain antibody directed against a therapeutic target is directed against any of GSK1, Bad, caspase and Forkhead.
    48. 48. A polypeptide construct according to item 47 for use in treating cancer.
    49. 49. Method for delivering an anti-target therapeutic compound to the interior of a cell comprising administering to a subject a polypeptide construct according to any of items 40 to 48.
    50. 50. Method for delivering an anti-target therapeutic compound to the interior of a cell without being inactivated comprising administering to a subject a polypeptide construct according to any of items 40 to 49.
    51. 51. A method according to item 50 wherein said cell is located in the gut system, and said a polypeptide construct is delivered orally.
    52. 52. A method according to item 50 wherein said cell is located in vaginal and/or rectal tract, and said a polypeptide construct is delivered to the vaginal and/or rectal tract.
    53. 53. A method according to item 50 wherein said cell is located in nose, upper respiratory tract and/or lung, and said a polypeptide construct is delivered to nose, upper respiratory tract and/or lung.
    54. 54. A method according to item 50 wherein said cell is located in intestinal mucosa, and said a polypeptide construct is delivered orally.
    55. 55. A method according to item 50 wherein said cell is located in the tissues beneath the tongue, and said a polypeptide construct is delivered to the tissues beneath the tongue.
    56. 56. A method according to item 50 wherein said cell is located in the skin, and said a polypeptide construct is delivered topically.
    57. 57. A method according to item 50 wherein said cell is in, or accessible via the blood, and said a polypeptide construct is delivered orally, to the vaginal and/or rectal tract, nasally, by inhalation though the mouth or nose, to the tissues beneath the tongue, or topically.
    58. 58. A polypeptide construct according to any of items 1 to 7, 19 to 24, 40 to 48, or a method according to any of items 11 to 18, 25 to 39, 49 to 57, wherein the single domain antibodies are humanized Camelidae VHHs.
    59. 59. A polypeptide construct according to any of items 1 to 7, 19 to 24, 40 to 48, 58, or a method according to any of items 11 to 18, 25 to 39, 49 to 57, wherein said single domain antibody is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length single domain antibody.
    60. 60. A polypeptide construct according any of items 1 to 7, 19 to 24, 40 to 48, 58 and 59 or a method according to any of items 11 to 18, 25 to 39, 49 to 59, wherein the polypeptide construct is an homologous sequence, a functional portion, or a functional portion of an homologous sequence of the full length polypeptide construct.
    61. 61. A polypeptide construct according to any of items 1 to 7, 19 to 24, 40 to 48, 58 to 60 or a method according to any of items 11 to 18, 25 to 39, 49 to 60 wherein said single domain antibodies are Camelidae VHHs.
    62. 62. A nucleic acid encoding a polypeptide construct according to any of items 1 to 7, 19 to 24, 40 to 48, 58 to 61.
    63. 63. A composition comprising a polypeptide construct as defined in any of the preceding items, together with a pharmaceutical carrier.
    Figure imgb0095
    Figure imgb0096
    Figure imgb0097
    Figure imgb0098
    Figure imgb0099
    Figure imgb0100
    Figure imgb0101
    Figure imgb0102
    Figure imgb0103
    Figure imgb0104
    Figure imgb0105
    Figure imgb0106
    Figure imgb0107
    Figure imgb0108
    Figure imgb0109
    Figure imgb0110
    Figure imgb0111
    Figure imgb0112
    Figure imgb0113
    Figure imgb0114
    Figure imgb0115
    Figure imgb0116
    Figure imgb0117
    Figure imgb0118
    Figure imgb0119
    Figure imgb0120
    Figure imgb0121
    Figure imgb0122
    Figure imgb0123
    Figure imgb0124
    Figure imgb0125
    Figure imgb0126
    Figure imgb0127
    Figure imgb0128
    Figure imgb0129
    Figure imgb0130
    Figure imgb0131
    Figure imgb0132
    Figure imgb0133
    Figure imgb0134
    Figure imgb0135
    Figure imgb0136
    Figure imgb0137
    Figure imgb0138
    Figure imgb0139
    Figure imgb0140
    Figure imgb0141
    Figure imgb0142
    Figure imgb0143
    Figure imgb0144
    Figure imgb0145
    Figure imgb0146
    Figure imgb0147
    Figure imgb0148
    Figure imgb0149
    Figure imgb0150
    Figure imgb0151
    Figure imgb0152
    Figure imgb0153
    Figure imgb0154
    Figure imgb0155
    Figure imgb0156
    Figure imgb0157
    Figure imgb0158
    Figure imgb0159
    Figure imgb0160
    Figure imgb0161
    Figure imgb0162
    Figure imgb0163
    Figure imgb0164

Claims (15)

  1. Composition for nasal administration comprising a therapeutically effective amount of a polypeptide construct comprising one or more single domain antibodies directed to one or more target molecule(s) and a pharmaceutically acceptable ingredient.
  2. Composition according to claim 1, wherein said pharmaceutically acceptable ingredient facilitates delivery of said polypeptide construct to the nose, the upper respiratory tract and/or lung or the bloodstream.
  3. Composition according to any of claims 1 or 2, which is in the form of a nasal spray or inhaler.
  4. Composition according to claim 3, which is an aerosol.
  5. Inhalation device comprising the composition according to any of claims 1 to 4 in the form of a dry powder.
  6. Composition according to any of claims 1 to 4, wherein, upon nasal administration of said composition to a subject, said polypeptide construct is delivered to the nose, upper respiratory tract and/or lung without being inactivated.
  7. Composition according to any of claim 1 to 4, wherein, upon nasal administration of said composition to a subject, said polypeptide construct is delivered to the bloodstream without being inactivated.
  8. Composition according to any of claims 1 to 7, wherein the target molecule against which said polypeptide construct is used, is TNF.
  9. Composition according to claim 8, wherein said polypeptide construct comprises at least one single domain antibody directed against TNF-alpha and corresponding to a sequence represented by SEQ ID NOs: 12 to 14.
  10. Composition according to any of claims 1 to 7, wherein the polypeptide construct comprises at least one single domain antibody directed against EGFR.
  11. Composition according to claim 12, wherein said at least one single domain antibody directed against EGF corresponds to a sequence represented by any of SEC2 ID NOs: 23 to 44.
  12. Composition according to any of claims 1 to 7, wherein the polypeptide construct comprises at least one single domain antibody directed against IgE,
  13. Composition according to claim 16, wherein said at least one single domain antibody directed against IgE corresponds to a sequence represented by any of SEQ ID NC3s: 1 to 11.
  14. Composition according to any of claims 1 to 7, wherein the polypeptide construct comprises at least one single domain antibody directed against MMP-12.
  15. Composition according to claim 19, wherein said at least one single domain antibody directed against MMP-12 corresponds to a sequence represented by any of SEQ ID NOs: 15 to 22.
EP10178270A 2002-11-08 2003-11-07 Single domain antibodies for nasal administration Withdrawn EP2301967A3 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP10178270A EP2301967A3 (en) 2002-11-08 2003-11-07 Single domain antibodies for nasal administration

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US42506302P 2002-11-08 2002-11-08
US42507302P 2002-11-08 2002-11-08
EP03447005 2003-01-10
EPPCT/EP03/06581 2003-06-23
EPPCT/EP03/07313 2003-07-08
EP10178270A EP2301967A3 (en) 2002-11-08 2003-11-07 Single domain antibodies for nasal administration
EP03775004A EP1558650A2 (en) 2002-11-08 2003-11-07 Camelidae antibodies against immunoglobulin e and use thereof for the treatment of allergic disorders

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
EP03775004.9 Division 2003-11-07

Publications (2)

Publication Number Publication Date
EP2301967A2 true EP2301967A2 (en) 2011-03-30
EP2301967A3 EP2301967A3 (en) 2011-07-13

Family

ID=32314976

Family Applications (8)

Application Number Title Priority Date Filing Date
EP10183400A Withdrawn EP2336179A1 (en) 2002-11-08 2003-11-07 Stabilized single domain antibodies
EP10182757.4A Expired - Lifetime EP2390268B1 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
EP10178264A Withdrawn EP2258392A1 (en) 2002-11-08 2003-11-07 Method of administering therapeutic polypeptides
EP10183379.6A Expired - Lifetime EP2316852B1 (en) 2002-11-08 2003-11-07 Stabilized single domain antibodies
EP17186411.9A Withdrawn EP3299393A1 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
EP10178263A Withdrawn EP2284192A3 (en) 2002-11-08 2003-11-07 Camelidae antibodies for sublingual administration
EP07014054.6A Expired - Lifetime EP1900753B1 (en) 2002-11-08 2003-11-07 Method of administering therapeutic polypeptides, and polypeptides therefor
EP10178270A Withdrawn EP2301967A3 (en) 2002-11-08 2003-11-07 Single domain antibodies for nasal administration

Family Applications Before (7)

Application Number Title Priority Date Filing Date
EP10183400A Withdrawn EP2336179A1 (en) 2002-11-08 2003-11-07 Stabilized single domain antibodies
EP10182757.4A Expired - Lifetime EP2390268B1 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
EP10178264A Withdrawn EP2258392A1 (en) 2002-11-08 2003-11-07 Method of administering therapeutic polypeptides
EP10183379.6A Expired - Lifetime EP2316852B1 (en) 2002-11-08 2003-11-07 Stabilized single domain antibodies
EP17186411.9A Withdrawn EP3299393A1 (en) 2002-11-08 2003-11-07 Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
EP10178263A Withdrawn EP2284192A3 (en) 2002-11-08 2003-11-07 Camelidae antibodies for sublingual administration
EP07014054.6A Expired - Lifetime EP1900753B1 (en) 2002-11-08 2003-11-07 Method of administering therapeutic polypeptides, and polypeptides therefor

Country Status (11)

Country Link
US (7) US20060115470A1 (en)
EP (8) EP2336179A1 (en)
JP (5) JP2006519763A (en)
AU (1) AU2010226938B2 (en)
CA (3) CA2505325C (en)
HK (1) HK1082745A1 (en)
IL (1) IL168357A (en)
MX (1) MXPA05004955A (en)
NZ (4) NZ540194A (en)
RU (1) RU2704232C2 (en)
WO (3) WO2004041863A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2530553C2 (en) * 2012-11-07 2014-10-10 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Московский государственный университет имени М.В. Ломоносова" (МГУ) Recombinant single-domain antibody able to bind human tumour necrosis factor specifically and its derivatives

Families Citing this family (367)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060073141A1 (en) * 2001-06-28 2006-04-06 Domantis Limited Compositions and methods for treating inflammatory disorders
JP2005289809A (en) 2001-10-24 2005-10-20 Vlaams Interuniversitair Inst Voor Biotechnologie Vzw (Vib Vzw) Mutant heavy-chain antibody
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
US20060002935A1 (en) * 2002-06-28 2006-01-05 Domantis Limited Tumor Necrosis Factor Receptor 1 antagonists and methods of use therefor
CN1678634A (en) 2002-06-28 2005-10-05 多曼蒂斯有限公司 Immunoglobulin single variable antigen combination area and its opposite constituent
US9028822B2 (en) 2002-06-28 2015-05-12 Domantis Limited Antagonists against TNFR1 and methods of use therefor
ATE512364T1 (en) 2002-08-07 2011-06-15 Ablynx Nv MODULATION OF PLATELE ADHESION BASED ON THE SURFACE-EXPOSED BETA-SWITCH LOOP OF THE PLATELET GLYCOPROTEIN IB-ALPHA
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
AU2003286004A1 (en) * 2002-11-08 2004-06-07 Ablynx N.V. Single domain antibodies directed against interferon- gamma and uses therefor
EP2336179A1 (en) * 2002-11-08 2011-06-22 Ablynx N.V. Stabilized single domain antibodies
ES2466716T3 (en) * 2002-11-08 2014-06-11 Ablynx N.V. Stabilized single domain antibodies
US20100003253A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US20060034833A1 (en) * 2002-11-08 2006-02-16 Els Beirnaert Single domain antibodies directed against interferron-gamma and uses therefor
US20060034845A1 (en) * 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US20060228355A1 (en) * 2003-11-07 2006-10-12 Toon Laeremans Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
AU2004204262B2 (en) 2003-01-10 2010-11-04 Ablynx N.V. Recombinant VHH single domain antibody from camelidae against von willebrand factor (vWF) or against collagen
PT1639011E (en) 2003-06-30 2009-01-20 Domantis Ltd Pegylated single domain antibodies (dab)
JP2008500830A (en) * 2004-06-01 2008-01-17 ドマンティス リミテッド Bispecific fusion antibodies with increased serum half-life
JP2008504356A (en) * 2004-06-30 2008-02-14 ドマンティス リミテッド Compositions and methods for treating inflammatory diseases
US8603824B2 (en) 2004-07-26 2013-12-10 Pfenex, Inc. Process for improved protein expression by strain engineering
US7563443B2 (en) 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
RU2401842C2 (en) 2004-10-08 2010-10-20 Домантис Лимитед Antagonists and method of using said antagonists
JP5113523B2 (en) * 2004-10-13 2013-01-09 アブリンクス ナームローゼ フェンノートシャップ Polypeptides comprising nanoantibodies against amyloid-β and nanoantibodies TM for the treatment of neurodegenerative diseases such as Alzheimer's disease
JP2008521870A (en) * 2004-12-02 2008-06-26 ドマンティス リミテッド Anti-IL-1R1 single domain antibody and therapeutic use
CA2589839C (en) * 2004-12-02 2016-04-05 Unilever N.V. Method for affinity purification
WO2006059106A2 (en) * 2004-12-02 2006-06-08 Domantis Limited Bispecific domain antibodies targeting serum albumin and glp-1 or pyy
EP2024396A2 (en) * 2004-12-02 2009-02-18 Domantis Limited Plad domain peptides with increased serum half life due to conjugation to domain antibodies
EP1772465B1 (en) 2005-01-05 2009-02-18 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Synthetic immunoglobulin domains with binding properties engineered in regions of the molecule different from the complementarity determining regions
ATE473446T1 (en) 2005-01-14 2010-07-15 Ablynx Nv METHODS AND TEST DEVICES FOR DIFFERENTIATING DIFFERENT FORMS OF DISEASES AND DISEASES CHARACTERIZED BY THROMBOCYTOPENIA AND/OR BY SPONTANEOUS INTERACTIONS BETWEEN THE VON WILLEBRAND FACTOR AND PLATES
MX2007011356A (en) * 2005-03-18 2008-03-10 Domantis Ltd Antibodies against candida antigens.
CN103254309B (en) * 2005-05-18 2017-09-26 埃博灵克斯股份有限公司 For the improved nanometer body of tumor necrosis factor αTM
AU2012200682B2 (en) * 2005-05-18 2014-11-27 Ablynx Nv Improved NanobodiesTM against Tumor Necrosis Factor-alpha
CN101213214B (en) 2005-05-20 2014-06-25 埃博灵克斯股份有限公司 Single domain VHH antibodies against von willebrand factor
DE102005023617A1 (en) * 2005-05-21 2006-11-23 Aspre Ag Method for mixing colors in a display
CA2619748C (en) 2005-08-30 2016-06-14 Actogenix Nv Novel treatment of chronic enterocolitis
US20090130776A1 (en) * 2005-09-01 2009-05-21 Canon Kabushiki Kaisha Binding protein molecule
WO2007035092A2 (en) * 2005-09-23 2007-03-29 Academisch Ziekenhuis Leiden Vhh for the diagnosis, prevention and treatment of diseases associated with protein aggregates
EP1940880A2 (en) * 2005-10-14 2008-07-09 Novo Nordisk A/S Treating diabetes using inhibitors of il-1
TW200736277A (en) 2005-11-14 2007-10-01 Amgen Inc RANKL antibody-PTH/PTHrP chimeric molecules
JP2007172129A (en) * 2005-12-20 2007-07-05 Sony Corp Nonvolatile memory access control device and nonvolatile memory control system
US20110059101A9 (en) * 2006-03-13 2011-03-10 Ablynx N.V. Amino Acid Sequences Directed Against Il-6 And Polypetides Comprising The Same For The Treatment Of Diseases And Disorders Associated With Il-6 Mediated Signalling
EP1999147A1 (en) * 2006-03-27 2008-12-10 Ablynx N.V. Medical delivery device for therapeutic proteins based on single domain antibodies
WO2007112940A2 (en) * 2006-03-31 2007-10-11 Ablynx N.V. Albumin-derived amino acid sequence, use thereof for increasing the half-life of therapeutic proteins and of other therapeutic compounds and entities, and constructs comprising the same
EP2010568A1 (en) * 2006-04-14 2009-01-07 Ablynx N.V. Dp-78-like nanobodies
AT503902B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING IMMUNE LOBULINS
AT503889B1 (en) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F MULTIVALENT IMMUNE LOBULINE
US20100113339A1 (en) * 2006-09-08 2010-05-06 Ablynx N. V. Serum albumin binding proteins with long half-lives
DK2698166T3 (en) 2006-10-10 2015-12-21 Regenesance B V KOMPLEMENTÆRHÆMNING FOR BETTER nerves GENERATION
AU2007306340A1 (en) * 2006-10-11 2008-04-17 Ablynx N.V. Amino acid sequences that bind to serum proteins in a manner that is essentially independent of the pH, compounds comprising the same, and use thereof
WO2008049897A1 (en) * 2006-10-27 2008-05-02 Ablynx N.V. Intranasal delivery of polypeptides and proteins
GB0621513D0 (en) 2006-10-30 2006-12-06 Domantis Ltd Novel polypeptides and uses thereof
WO2008068280A1 (en) * 2006-12-05 2008-06-12 Ablynx N.V. Peptides capable of binding to serum proteins
US20100303777A1 (en) * 2006-12-14 2010-12-02 Actogenix N.V. Delivery of binding molecules to induce immunomodulation
AU2007336243B2 (en) 2006-12-19 2012-07-26 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the ADAM family and polypeptides comprising the same for the treatment of ADAM-related diseases and disorders
EP2557090A3 (en) 2006-12-19 2013-05-29 Ablynx N.V. Amino acid sequences directed against GPCRs and polypeptides comprising the same for the treatment of GPCR-related diseases and disorders
PL2308514T3 (en) 2007-03-23 2013-11-29 To Bbb Holding B V Conjugates for targeted drug delivery across the blood-brain barrier
WO2008124858A2 (en) * 2007-04-11 2008-10-23 F-Star Biotechnologische Forschungs- Und Entwicklungsges. M.B.H. Targeted receptor
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
CN101796072B (en) 2007-05-24 2014-09-24 埃博灵克斯股份有限公司 Amino acid sequences directed against RANK-l and polypeptides comprising the same for the treatment of bone diseases and disorders
EP2164517A4 (en) * 2007-05-29 2011-03-09 Univ Yale Il- 18 and protein kinase r inhibition for the treatment of copd
CA2683791A1 (en) 2007-06-06 2008-12-11 Domantis Limited Polypeptides, antibody variable domains & antagonists
JP5602625B2 (en) 2007-06-26 2014-10-08 エフ−スター ビオテヒノロギッシェ フォルシュングス− ウント エントヴィッケルングスゲゼルシャフト ミット ベシュレンクテル ハフツング Binding substance display
US10214588B2 (en) 2007-07-03 2019-02-26 Ablynx N.V. Providing improved immunoglobulin sequences by mutating CDR and/or FR positions
EP2535351A3 (en) * 2007-09-26 2013-04-03 UCB Pharma S.A. Dual specificity antibody fusions
GB2453589A (en) 2007-10-12 2009-04-15 King S College London Protease inhibition
CA2706200A1 (en) * 2007-11-27 2009-06-04 Ablynx N.V. Immunoglobulin constructs comprising multiple single variable domains and an fc portion
WO2009067820A1 (en) 2007-11-27 2009-06-04 The University Of British Columbia 14-3-3 antagonists for the prevention and treatment of arthritis
EA201000785A1 (en) * 2007-12-13 2011-02-28 Глаксо Груп Лимитед COMPOSITIONS FOR DELIVERY IN LIGHT
EP2080770A1 (en) * 2008-01-21 2009-07-22 MorphoSys AG Proteinaceous binding molecules comprising purification tags
AU2009221915A1 (en) * 2008-03-03 2009-09-11 Dyax Corp. Metalloproteinase 12 binding proteins
JP2011525476A (en) 2008-03-05 2011-09-22 アブリンクス エン.ヴェー. Novel antigen-binding dimer complex, its production method and use
GB0805608D0 (en) * 2008-03-28 2008-04-30 Sec Dep For Environment Food & Detection method
GB0809069D0 (en) 2008-05-19 2008-06-25 Univ Leuven Kath Gene signatures
EP2947097A1 (en) 2008-04-07 2015-11-25 Ablynx N.V. Amino acid sequences directed against the Notch pathways and uses thereof
WO2009127691A1 (en) 2008-04-17 2009-10-22 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
EP2113255A1 (en) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Cytotoxic immunoglobulin
ES2620285T3 (en) 2008-05-02 2017-06-28 Novartis Ag Binding molecules based on improved fibronectin and their uses
US8444976B2 (en) 2008-07-02 2013-05-21 Argen-X B.V. Antigen binding polypeptides
KR101791372B1 (en) 2008-07-18 2017-10-27 브리스톨-마이어스 스큅 컴퍼니 Compositions monovalent for cd28 binding and methods of use
CN102164965B (en) 2008-09-26 2016-03-30 Ucb医药有限公司 Biological product
CA2740561C (en) * 2008-10-14 2021-01-19 National Research Council Of Canada Bsa-specific antibodies
MX2011004244A (en) * 2008-10-21 2011-05-25 Domantis Ltd Ligands that have binding specificity for dc-sign.
AU2013202856B2 (en) * 2008-10-29 2016-02-11 Ablynx N.V. Formulations of single domain antigen binding molecules
CN102272154A (en) 2008-10-29 2011-12-07 惠氏有限责任公司 Methods for purification of single domain antigen binding molecules
AU2009333791B2 (en) 2008-10-29 2013-04-04 Ablynx N.V. Formulations of single domain antigen binding molecules
KR101740066B1 (en) 2008-12-05 2017-05-25 글락소 그룹 리미티드 Methods for selecting protease resistant polypeptides
DK2387583T3 (en) * 2009-01-14 2019-01-14 Ablynx Nv PULMONAL ADMINISTRATION OF VARIABLE IMMUNGLOBULIN SINGLE DOMAINS AND CONSTRUCTIONS THEREOF.
KR20110119806A (en) 2009-02-19 2011-11-02 글락소 그룹 리미티드 Improved anti-tnfr1 polypeptides, antibody variable domains & antagonists
US9265834B2 (en) 2009-03-05 2016-02-23 Ablynx N.V. Stable formulations of polypeptides and uses thereof
WO2010100135A1 (en) 2009-03-05 2010-09-10 Ablynx N.V. Novel antigen binding dimer-complexes, methods of making/avoiding and uses thereof
US8748581B2 (en) 2009-04-10 2014-06-10 Ablynx N.V. Anti-IL-6R polypeptides and pharmaceutical compositions thereof
WO2010125187A2 (en) 2009-04-30 2010-11-04 Ablynx Nv Method for the production of domain antibodies
WO2010139808A2 (en) 2009-06-05 2010-12-09 Ablynx Nv IMPROVED AMINO ACID SEQUENCES DIRECTED AGAINST HUMAN RESPIRATORY SYNCYTIAL VIRUS (hRSV) AND POLYPEPTIDES COMPRISING THE SAME FOR THE PREVENTION AND/OR TREATMENT OF RESPIRATORY TRACT INFECTIONS
US9150640B2 (en) 2009-07-10 2015-10-06 Ablynx N.V. Method for the production of variable domains
US20120107330A1 (en) 2009-07-16 2012-05-03 Adriaan Allart Stoop Antagonists, uses & methods for partially inhibiting tnfr1
CN102548466A (en) 2009-07-28 2012-07-04 霍夫曼-拉罗奇有限公司 Non-invasive in vivo optical imaging method
CN102574922A (en) 2009-07-29 2012-07-11 葛兰素集团有限公司 Anti - TGF - BETA receptor type II single domain antibodies
LT2805731T (en) 2009-09-03 2019-02-11 Ablynx N.V. Stable formulations of polypeptides and uses thereof
GB201005063D0 (en) 2010-03-25 2010-05-12 Ucb Pharma Sa Biological products
UY32917A (en) 2009-10-02 2011-04-29 Boehringer Ingelheim Int DLL-4 BINDING MOLECULES
US20110172398A1 (en) 2009-10-02 2011-07-14 Boehringer Ingelheim International Gmbh Bispecific binding molecules for anti-angiogenesis therapy
EP2486056A1 (en) 2009-10-09 2012-08-15 Ablynx N.V. Immunoglobulin single variable domain directed against human cxcr4 and other cell associated proteins and methods to generate them
DK2491056T3 (en) * 2009-10-22 2021-10-25 Univ Twente VHH FOR USE IN TISSUE REPAIR, ORGAN REGENERATION, ORGAN REPLACEMENT AND TISSUE CONSTRUCTION
SG10201500274TA (en) 2009-10-27 2015-03-30 Glaxo Group Ltd Stable anti-tnfr1 polypeptides, antibody variable domains & antagonists
ES2691717T3 (en) 2009-10-30 2018-11-28 Novartis Ag Universal libraries of the binding domain of the inferior side of type III fibronectin type III
WO2011051327A2 (en) 2009-10-30 2011-05-05 Novartis Ag Small antibody-like single chain proteins
EP2507262A1 (en) 2009-11-30 2012-10-10 Ablynx N.V. Improved amino acid sequences directed against human respiratory syncytial virus (hrsv) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
EP3309176A1 (en) 2009-12-14 2018-04-18 Ablynx N.V. Immunoglobulin single variable domain antibodies against ox40l, constructs and therapeutic use
WO2011083141A2 (en) 2010-01-08 2011-07-14 Ablynx Nv Method for generation of immunoglobulin sequences by using lipoprotein particles
TWI513466B (en) 2010-01-20 2015-12-21 Boehringer Ingelheim Int Anticoagulant antidotes
CN102781959A (en) 2010-02-05 2012-11-14 埃博灵克斯股份有限公司 Peptides capable of binding to serum albumin and compounds, constructs and polypeptides comprising the same
US9120855B2 (en) 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
EP2533814A2 (en) 2010-02-11 2012-12-19 Ablynx N.V. Delivery of immunoglobulin variable domains and constructs thereof
CN102753148B (en) * 2010-02-11 2018-01-26 埃博灵克斯股份有限公司 For preparing the method and composition of aerosol
TW201144437A (en) 2010-03-03 2011-12-16 Boehringer Ingelheim Int A-beta binding polypeptides
CA2794551A1 (en) 2010-03-26 2011-09-29 Westfaelische Wilhelms-Universitaet Muenster Substitute therapy for glucocorticoids
WO2011117423A1 (en) 2010-03-26 2011-09-29 Ablynx N.V. Immunoglobulin single variable domains directed against cxcr7
US9101674B2 (en) 2010-03-29 2015-08-11 Vib Vzw Targeting and in vivo imaging of tumor-associated macrophages
US9556273B2 (en) 2010-03-29 2017-01-31 Vib Vzw Anti-macrophage mannose receptor single variable domains for targeting and in vivo imaging of tumor-associated macrophages
EP4234698A3 (en) 2010-05-06 2023-11-08 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
KR20130036012A (en) 2010-05-07 2013-04-09 에프. 호프만-라 로슈 아게 Diagnostic method for the detection of cells ex vivo
KR20130080790A (en) 2010-05-20 2013-07-15 아블린쓰 엔.브이. Biological materials related to her3
WO2011161263A1 (en) 2010-06-25 2011-12-29 Ablynx Nv Pharmaceutical compositions for cutaneous administration
WO2012007880A2 (en) 2010-07-16 2012-01-19 Ablynx Nv Modified single domain antigen binding molecules and uses thereof
EP3248987A1 (en) 2010-07-30 2017-11-29 Novartis AG Fibronectin cradle molecules and libraries thereof
MY162825A (en) 2010-08-20 2017-07-31 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
US20120225081A1 (en) 2010-09-03 2012-09-06 Boehringer Ingelheim International Gmbh Vegf-binding molecules
US20120244141A1 (en) 2010-09-28 2012-09-27 Boehringer Ingelheim International Gmbh Stratification of cancer patients for susceptibility to therapy with PTK2 inhibitors
US11644471B2 (en) 2010-09-30 2023-05-09 Ablynx N.V. Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains
EP2621953B1 (en) 2010-09-30 2017-04-05 Ablynx N.V. Biological materials related to c-met
NO2632946T3 (en) 2010-10-29 2018-05-05
JP5933573B2 (en) 2010-11-08 2016-06-15 ノバルティス アーゲー CXCR2 binding polypeptide
BR112013012786B1 (en) 2010-11-26 2022-02-08 Molecular Partners Ag RECOMBINANT PROTEIN COMPRISING AN ANCHYRIN REPEAT DOMAIN AND PHARMACEUTICAL COMPOSITION
EP2661449B1 (en) 2011-01-06 2017-03-22 Glaxo Group Limited Ligands that bind tgf-beta receptor ii
JP6385060B2 (en) 2011-03-07 2018-09-05 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト In vivo selection of therapeutically active antibodies
EP2683290B1 (en) 2011-03-07 2018-11-07 F.Hoffmann-La Roche Ag Methods for in vivo testing of therapeutic antibodies
AU2012234282B2 (en) 2011-03-28 2015-07-16 Ablynx Nv Method for producing solid formulations comprising immunoglobulin single variable domains
US20150158948A9 (en) 2011-03-28 2015-06-11 Francis Descamps Bispecific anti-cxcr7 immunoglobulin single variable domains
MA34978B1 (en) 2011-03-30 2014-03-01 Boehringer Ingelheim Int ANTIDOTES FOR ANTICOAGULANTS
WO2012131053A1 (en) 2011-03-30 2012-10-04 Ablynx Nv Methods of treating immune disorders with single domain antibodies against tnf-alpha
US20130078247A1 (en) 2011-04-01 2013-03-28 Boehringer Ingelheim International Gmbh Bispecific binding molecules binding to dii4 and ang2
US9527925B2 (en) 2011-04-01 2016-12-27 Boehringer Ingelheim International Gmbh Bispecific binding molecules binding to VEGF and ANG2
UA117218C2 (en) 2011-05-05 2018-07-10 Мерк Патент Гмбх Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
EP2707382B1 (en) 2011-05-09 2019-07-17 Ablynx NV Method for the production of immunoglobulin single variable domains
EP2714736A1 (en) 2011-05-27 2014-04-09 Ablynx N.V. Inhibition of bone resorption with rankl binding peptides
US9580480B2 (en) 2011-05-31 2017-02-28 Massachusetts Institute Of Technology Cell-directed synthesis of multifunctional nanopatterns and nanomaterials
AU2012273928A1 (en) 2011-06-23 2014-02-06 Ablynx Nv Immunoglobulin single variable domains directed against IgE
JP6324887B2 (en) 2011-06-23 2018-05-16 アブリンクス エン.ヴェー. Proteins that bind to serum albumin
MX350074B (en) 2011-06-23 2017-08-25 Ablynx Nv Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains.
EP3839049A3 (en) 2011-09-19 2021-10-20 Kymab Limited Antibodies, variable domains & chains tailored for human use
DK2747782T3 (en) 2011-09-23 2018-04-23 Ablynx Nv Long-term inhibition of interleukin-6-mediated signal transmission
EP2761008A1 (en) 2011-09-26 2014-08-06 Kymab Limited Chimaeric surrogate light chains (slc) comprising human vpreb
US9346884B2 (en) 2011-09-30 2016-05-24 Ablynx N.V. Biological materials related to c-Met
CN103889451B (en) 2011-09-30 2016-06-29 埃博灵克斯股份有限公司 The biological substance relevant to C-Met
GB2496375A (en) 2011-10-28 2013-05-15 Kymab Ltd A non-human assay vertebrate comprising human antibody loci and human epitope knock-in, and uses thereof
CN104039830A (en) 2011-11-04 2014-09-10 诺华股份有限公司 Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
GB201122047D0 (en) 2011-12-21 2012-02-01 Kymab Ltd Transgenic animals
KR102151383B1 (en) 2011-12-05 2020-09-03 노파르티스 아게 Antibodies for epidermal growth factor receptor 3 (her3)
EA201491107A1 (en) 2011-12-05 2014-11-28 Новартис Аг ANTIBODIES TO THE RECEPTOR EPIDERMAL GROWTH FACTOR 3 (HER3), DIRECTED TO DOMAIN II HER3
JP2015504064A (en) * 2012-01-06 2015-02-05 コンプリクス エン ヴェー Binding agents for intracellular target molecules
PL2831111T3 (en) 2012-03-30 2019-09-30 Boehringer Ingelheim International Gmbh Ang2-binding molecules
US9328174B2 (en) 2012-05-09 2016-05-03 Novartis Ag Chemokine receptor binding polypeptides
US9289507B2 (en) 2012-05-17 2016-03-22 Extend Biosciences, Inc. Carriers for improved drug delivery
JP6411333B2 (en) 2012-05-24 2018-10-24 ブイアイビー ブイゼットダブリュVib Vzw Anti-macrophage mannose receptor single variable domain for tumor-associated macrophage targeting and INVIVO imaging
US11339208B1 (en) 2012-05-31 2022-05-24 United States Of America As Represented By The Secretary Of The Air Force Camelidae single-domain antibodies against Yersinia pestis and methods of use
DK2890711T3 (en) * 2012-08-31 2017-02-27 Argen-X Nv Process for Preparation of Inter-species, Intra-Target Cross-Reactivity Antibody Molecules
EP2895510A2 (en) * 2012-09-13 2015-07-22 Novartis AG Single domain antibody with c-terminal modification
WO2014087010A1 (en) 2012-12-07 2014-06-12 Ablynx N.V. IMPROVED POLYPEPTIDES DIRECTED AGAINST IgE
EP2752426A1 (en) 2013-01-03 2014-07-09 Covagen AG Human serum albumin binding compounds and fusion proteins thereof
WO2014111550A1 (en) 2013-01-17 2014-07-24 Glaxosmithkline Intellectual Property Development Limited Modified anti-serum albumin binding proteins
ES2654675T3 (en) 2013-01-30 2018-02-14 Vib Vzw Novel chimeric polypeptides for drug testing and discovery purposes
WO2014120916A1 (en) 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Pegylated domain antibodies monovalent for cd28 binding and methods of use
ES2745772T3 (en) 2013-02-05 2020-03-03 Vib Vzw Acetylcholine Muscarinic Receptor Binding Agents and Uses thereof
WO2014165093A2 (en) 2013-03-13 2014-10-09 Bristol-Myers Squibb Company Fibronectin based scaffold domains linked to serum albumin or a moiety binding thereto
EP2970479B1 (en) 2013-03-14 2019-04-24 Novartis AG Antibodies against notch 3
AU2014229952B2 (en) 2013-03-15 2018-10-04 Vib Vzw Anti-macrophage mannose receptor single variable domains for use in cardiovascular diseases
CN105358699A (en) 2013-04-29 2016-02-24 阿格罗塞文公司 Agrochemical compositions comprising antibodies binding to sphingolipids
WO2016071438A2 (en) 2014-11-05 2016-05-12 Agrosavfe Nv Transgenic plant comprising a polynucleotide encoding a variable domain of heavy-chain antibody
NL1040254C2 (en) 2013-05-17 2014-11-24 Ablynx Nv Stable formulations of immunoglobulin single variable domains and uses thereof.
JP6449272B2 (en) 2013-07-08 2019-01-09 ナンジン レジェンド バイオテック カンパニー,リミテッドNanjing Legend Biotech Co.,Ltd. Compositions and methods for increasing the serum half-life of proteins
CN103454413B (en) * 2013-09-04 2015-03-11 吉日木图 Bactrian camel specific antibody preparation method and immune detection method
EP2883883A1 (en) 2013-12-16 2015-06-17 Cardio3 Biosciences S.A. Therapeutic targets and agents useful in treating ischemia reperfusion injury
EP3087101A4 (en) 2013-12-20 2017-12-06 Novartis AG Regulatable chimeric antigen receptor
EP3119425B1 (en) 2014-03-15 2020-09-23 Novartis AG Regulatable chimeric antigen receptor
CN109053885A (en) 2014-05-16 2018-12-21 埃博灵克斯股份有限公司 Improved immunoglobulin variable domain domain
ES2900852T3 (en) 2014-05-16 2022-03-18 Ablynx Nv Methods of detection and/or measurement of anti-drug antibodies, in particular anti-drug antibodies that occur during treatment
DK3143042T3 (en) 2014-05-16 2020-08-31 Ablynx Nv IMPROVED VARIABLE IMMUNOGLOBULIN DOMAINS
NL2013661B1 (en) 2014-10-21 2016-10-05 Ablynx Nv KV1.3 Binding immunoglobulins.
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016012363A1 (en) 2014-07-22 2016-01-28 Vib Vzw Methods to select for agents that stabilize protein complexes
WO2016016329A1 (en) 2014-07-29 2016-02-04 Vrije Universiteit Brussel Radio-labelled antibody fragments for use in the prognosis, diagnosis of cancer as well as for the prediction of cancer therapy response
CN106573983B (en) 2014-07-29 2018-05-25 布鲁塞尔自由大学 For the radiolabeled antibody fragment used in the prevention and/or treatment of cancer
EP3174546B1 (en) 2014-07-31 2019-10-30 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
CA2958578A1 (en) 2014-08-21 2016-02-25 The General Hospital Corporation Tumor necrosis factor superfamily and tnf-like ligand muteins and methods of preparing and using the same
WO2016065042A1 (en) 2014-10-22 2016-04-28 Extend Biosciences, Inc. Therapeutic vitamin d conjugates
US9789197B2 (en) 2014-10-22 2017-10-17 Extend Biosciences, Inc. RNAi vitamin D conjugates
WO2016065052A1 (en) 2014-10-22 2016-04-28 Extend Biosciences, Inc. Insulin vitamin d conjugates
US20170267784A1 (en) 2014-10-23 2017-09-21 Singh Molecular Medicine, Llc Single domain antibodies directed against intracellular antigens
WO2016065323A2 (en) 2014-10-23 2016-04-28 Singh Biotechnology, Llc Single domain antibodies directed against intracellular antigens
WO2016097313A1 (en) 2014-12-19 2016-06-23 Ablynx N.V. Cysteine linked nanobody dimers
IL254577B2 (en) * 2015-03-31 2023-11-01 Vhsquared Ltd Polypeptides
EP4345112A2 (en) 2015-05-13 2024-04-03 Ablynx N.V. T cell recruiting polypeptides based on cd3 reactivity
RS59376B1 (en) 2015-05-13 2019-11-29 Ablynx Nv T cell recruiting polypeptides based on tcr alpha/beta reactivity
DK3298033T4 (en) 2015-05-18 2023-10-02 Tcr2 Therapeutics Inc COMPOSITIONS AND MEDICAL USES FOR TCR REPROGRAMMING USING FUSION PROTEINS
SG10202002131PA (en) 2015-05-21 2020-05-28 Harpoon Therapeutics Inc Trispecific binding proteins and methods of use
EP3325020B1 (en) 2015-07-17 2022-01-12 Vrije Universiteit Brussel Radiolabelled antibody fragments for use in treating cancer
BR112018001353A2 (en) 2015-08-07 2018-09-11 Alexo Therapeutics, Inc. constructs having a sirp-alpha domain or variant thereof
EA034582B1 (en) 2015-08-07 2020-02-21 АЭлЭкс ОНКОЛОДЖИ ИНК. Sirp-alpha variant constructs and use thereof
TWI746473B (en) 2015-11-02 2021-11-21 美商辛分子醫藥有限公司 Single domain antibodies directed against intracellular antigens
NO2768984T3 (en) 2015-11-12 2018-06-09
HUE057241T2 (en) 2015-11-13 2022-04-28 Ablynx Nv Improved serum albumin-binding immunoglobulin variable domains
US11077187B2 (en) * 2015-11-17 2021-08-03 Oklahoma Medical Research Foundation Epitope of optimized humanized monoclonal antibodies against activated protein C and uses thereof
GEP20217220B (en) 2015-11-18 2021-02-10 Merck Sharp & Dohme Pd1 and/or lag3 binders
US10501542B2 (en) 2015-11-18 2019-12-10 Merck Sharp & Dohme Corp. CTLA4 binders
JO3740B1 (en) 2015-11-18 2021-01-31 Merck Sharp & Dohme PD1/CTLA4 Binders
CA3005488A1 (en) 2015-11-18 2017-05-26 Ablynx Nv Improved serum albumin binders
MA43302A (en) 2015-11-27 2021-03-17 Ablynx Nv LIGAND CD40L INHIBITED POLYPEPTIDES
CA3009075A1 (en) * 2016-01-11 2017-07-20 Inhibrx, Inc. Multivalent and multispecific ox40-binding fusion proteins
CN109069627A (en) * 2016-01-14 2018-12-21 纪念斯隆-凯特琳癌症中心 To the T cell receptor sample antibody of the derivative peptide specific of FOXP3
CN108883180B (en) 2016-02-05 2023-07-07 奥里尼斯生物科学私人有限公司 CLEC9A binding agents and uses thereof
CA3016849A1 (en) 2016-03-07 2017-09-14 Vib Vzw Cd20 binding single domain antibodies
US11243214B2 (en) 2016-04-22 2022-02-08 Université Libre de Bruxelles Biomarker expressed in pancreatic beta cells useful in imaging or targeting beta cells
WO2017182605A1 (en) 2016-04-22 2017-10-26 Université Libre de Bruxelles A new biomarker expressed in pancreatic beta cells useful in imaging or targeting beta cells
JP2019523213A (en) 2016-05-02 2019-08-22 アブリンクス エン.ヴェー. Treatment of RSV infection
EP3455245A2 (en) 2016-05-13 2019-03-20 Orionis Biosciences NV Therapeutic targeting of non-cellular structures
KR20220103806A (en) 2016-05-18 2022-07-22 베링거 인겔하임 인터내셔날 게엠베하 Anti pd-1 and anti-lag3 antibodies for cancer treatment
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
CA3024723A1 (en) 2016-05-20 2017-11-23 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
CN116987189A (en) 2016-05-20 2023-11-03 哈普恩治疗公司 Single chain variable fragment CD3 binding proteins
PL3475699T3 (en) 2016-06-23 2022-09-26 Ablynx N.V. Improved pharmacokinetic assays for immunoglobulin single variable domains
WO2018007442A1 (en) 2016-07-06 2018-01-11 Ablynx N.V. Treatment of il-6r related diseases
AU2017306432A1 (en) 2016-08-02 2019-03-21 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018029182A1 (en) 2016-08-08 2018-02-15 Ablynx N.V. Il-6r single variable domain antibodies for treatment of il-6r related diseases
WO2018050833A1 (en) 2016-09-15 2018-03-22 Ablynx Nv Immunoglobulin single variable domains directed against macrophage migration inhibitory factor
EP3519438A1 (en) 2016-09-30 2019-08-07 VHsquared Limited Compositions
JP7217970B2 (en) 2016-10-07 2023-02-06 ティーシーアール2 セラピューティクス インク. Compositions and methods for reprogramming T-cell receptors using fusion proteins
CA3043515A1 (en) 2016-11-16 2018-05-24 Ablynx Nv T cell recruiting polypeptides capable of binding cd123 and tcr alpha/beta
WO2018098365A2 (en) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
EA201991168A1 (en) 2016-11-23 2019-12-30 Харпун Терапьютикс, Инк. PROTEIN BINDING PROSTATIC SPECIFIC MEMBRANE ANTIGEN
BR112019010602A2 (en) 2016-11-23 2019-12-17 Harpoon Therapeutics Inc trispecific psma proteins and methods of use
WO2018099968A1 (en) 2016-11-29 2018-06-07 Ablynx N.V. Treatment of infection by respiratory syncytial virus (rsv)
MX2019006574A (en) 2016-12-07 2019-08-21 Ablynx Nv Improved serum albumin binding immunoglobulin single variable domains.
EP3360898A1 (en) 2017-02-14 2018-08-15 Boehringer Ingelheim International GmbH Bispecific anti-tnf-related apoptosis-inducing ligand receptor 2 and anti-cadherin 17 binding molecules for the treatment of cancer
CN107674122A (en) * 2016-12-28 2018-02-09 天津天锐生物科技有限公司 A kind of single domain antibody for identifying human serum albumins
BR112019014600A2 (en) 2017-01-17 2020-02-18 Ablynx N.V. IMPROVED SERUM ALBUMIN BINDERS
AU2018209151A1 (en) 2017-01-17 2019-07-25 Ablynx Nv Improved serum albumin binders
GB201701404D0 (en) * 2017-01-27 2017-03-15 Micropharm Ltd Therapies for treating inflammatory disorders
WO2018141964A1 (en) 2017-02-06 2018-08-09 Orionis Biosciences Nv Targeted chimeric proteins and uses thereof
US11535668B2 (en) 2017-02-28 2022-12-27 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
CN110573604A (en) 2017-02-28 2019-12-13 非营利性组织佛兰芒综合大学生物技术研究所 Means and methods for oral protein delivery
CN110520436A (en) 2017-03-15 2019-11-29 潘迪恩治疗公司 Target immunological tolerance
US20200188541A1 (en) 2017-03-30 2020-06-18 Duke University Radiolabeled biomolecules and their use
AU2018241286A1 (en) 2017-03-31 2019-09-26 Ablynx N.V. Improved immunogenicity assays
US20200033347A1 (en) 2017-04-18 2020-01-30 Universite Libre De Bruxelles Biomarkers And Targets For Proliferative Diseases
KR20200005635A (en) 2017-05-11 2020-01-15 브이아이비 브이지더블유 Glycosylation of Variable Immunoglobulin Domains
WO2018209304A1 (en) 2017-05-12 2018-11-15 Harpoon Therapeutics, Inc. Msln targeting trispecific proteins and methods of use
CN113896792A (en) 2017-05-12 2022-01-07 哈普恩治疗公司 Mesothelin binding proteins
AU2018273914A1 (en) 2017-05-24 2019-11-14 Pandion Operations, Inc. Targeted immunotolerance
BR112019025392A2 (en) 2017-06-02 2020-07-07 Ablynx N.V. aggrecan-binding immunoglobulins
BR112019025097A2 (en) * 2017-06-02 2020-07-28 Merck Patent Gmbh mmp13-binding immunoglobulins
JP7249961B2 (en) 2017-06-02 2023-03-31 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Polypeptides that bind ADAMTS5, MMP13 and aggrecan
BR112019024333A2 (en) 2017-06-02 2020-07-28 Merck Patent Gmbh adamts binding immunoglobulins
GB201710973D0 (en) 2017-07-07 2017-08-23 Avacta Life Sciences Ltd Scaffold proteins
CN111201246B (en) 2017-07-11 2023-10-27 亚力兄制药公司 Polypeptide binding complement component C5 or serum albumin and fusion protein thereof
CA3070253A1 (en) 2017-07-19 2019-01-24 Vib Vzw Serum albumin binding agents
AU2018347582A1 (en) 2017-10-13 2020-05-07 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
MX2020003856A (en) 2017-10-13 2020-08-13 Harpoon Therapeutics Inc B cell maturation antigen binding proteins.
US11873347B2 (en) 2017-10-31 2024-01-16 Vib Vzw Antigen-binding chimeric proteins and methods and uses thereof
US10174092B1 (en) 2017-12-06 2019-01-08 Pandion Therapeutics, Inc. IL-2 muteins
US10946068B2 (en) 2017-12-06 2021-03-16 Pandion Operations, Inc. IL-2 muteins and uses thereof
CN109970860A (en) * 2017-12-27 2019-07-05 信达生物制药(苏州)有限公司 Three chain antibodies, Its Preparation Method And Use
KR20200128533A (en) 2018-02-05 2020-11-13 오리오니스 바이오사이언시즈 인코포레이티드 Fibroblast binding agents and uses thereof
WO2019155041A1 (en) 2018-02-12 2019-08-15 Vib Vzw Gβγ COMPLEX ANTIBODIES AND USES THEREOF
CA3092421A1 (en) 2018-03-01 2019-09-06 Vrije Universiteit Brussel Human pd-l1-binding immunoglobulins
AU2019237215B2 (en) 2018-03-23 2023-11-02 Université Libre de Bruxelles Wnt signaling agonist molecules
AU2019241439A1 (en) 2018-03-27 2020-10-22 Umc Utrecht Holding B.V. Targeted thrombolysis for treatment of microvascular thrombosis
SG11202011309SA (en) 2018-05-14 2020-12-30 Werewolf Therapeutics Inc Activatable interleukin 12 polypeptides and methods of use thereof
CA3100007A1 (en) 2018-05-14 2019-11-21 Werewolf Therapeutics, Inc. Activatable interleukin-2 polypeptides and methods of use thereof
EP3569618A1 (en) 2018-05-19 2019-11-20 Boehringer Ingelheim International GmbH Antagonizing cd73 antibody
JP7122672B2 (en) * 2018-06-08 2022-08-22 パナソニックIpマネジメント株式会社 VHH antibodies
US20210315933A1 (en) * 2018-07-26 2021-10-14 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using target specific fusion proteins
EP3837285A1 (en) 2018-08-13 2021-06-23 Inhibrx, Inc. Ox40-binding polypeptides and uses thereof
JP7190675B2 (en) * 2018-08-23 2022-12-16 パナソニックIpマネジメント株式会社 Antibody and complex that bind to norovirus, detection device and detection method using the same
JP7425049B2 (en) 2018-09-25 2024-01-30 ハープーン セラピューティクス,インク. DLL3 binding protein and method of use
WO2020069398A1 (en) 2018-09-27 2020-04-02 Akrevia Therapeutics Inc. Masked cytokine polypeptides
EP3636657A1 (en) 2018-10-08 2020-04-15 Ablynx N.V. Chromatography-free antibody purification method
GB201818477D0 (en) 2018-11-13 2018-12-26 Emstopa Ltd Tissue plasminogen activator antibodies and method of use thereof
WO2020114616A1 (en) * 2018-12-07 2020-06-11 Tillotts Pharma Ag Topical treatment of immune checkpoint inhibitor induced diarrhoea, colitis or enterocolitis using antibodies and fragments thereof
CN112830967B (en) * 2019-01-10 2022-06-24 瑞阳(苏州)生物科技有限公司 Affinity chromatography purification method of anti-HSA single domain antibody and fusion protein thereof
KR20220016077A (en) 2019-04-29 2022-02-08 콘포 테라퓨틱스 엔.브이. Chimeric proteins and methods for screening compounds and ligands that bind to GPCRs
US20220289837A1 (en) 2019-04-30 2022-09-15 Vib Vzw Cystic Fibrosis Transmembrane Conductance Regulator Stabilizing Agents
MX2021013766A (en) 2019-05-14 2022-02-21 Werewolf Therapeutics Inc Separation moieties and methods and use thereof.
KR20220035333A (en) 2019-05-20 2022-03-22 팬디온 오퍼레이션스, 인코포레이티드 MADCAM Targeted Immune Tolerance
EP3976650A1 (en) 2019-05-28 2022-04-06 Vib Vzw Cancer treatment by targeting plexins in the immune compartment
US20220228116A1 (en) 2019-05-28 2022-07-21 Vib Vzw Cd8+ t-cells lacking plexins and their application in cancer treatment
KR20220044904A (en) 2019-05-31 2022-04-12 알렉소 온콜로지 인크. How to treat cancer with SIRP alpha FC fusion in combination with immune checkpoint inhibitors
MX2021015762A (en) 2019-06-21 2022-04-18 Sorriso Pharmaceuticals Inc Compositions.
US20230056445A1 (en) 2019-06-21 2023-02-23 Sorriso Pharmaceuticals, Inc. Polypeptides
AU2020296979A1 (en) 2019-06-21 2022-02-24 Sorriso Pharmaceuticals, Inc. Polypeptides
TW202128775A (en) 2019-10-16 2021-08-01 英商阿法克塔生命科學有限公司 Pd-l1 inhibitor - tgfβ inhibitor bispecific drug moieties
WO2021078786A1 (en) 2019-10-21 2021-04-29 Vib Vzw Nanodisc-specific antigen-binding chimeric proteins
CA3160506A1 (en) 2019-11-11 2021-05-20 Ibi-Ag Innovative Bio Insecticides Ltd. Insect control nanobodies and uses thereof
US20220411495A1 (en) 2019-11-27 2022-12-29 Vib Vzw Positive allosteric modulators of the calcium-sensing receptor
WO2021110816A1 (en) 2019-12-06 2021-06-10 Ablynx Nv POLYPEPTIDES COMPRISING IMMUNOGLOBULIN SINGLE VARIABLE DOMAINS TARGETING TNFα AND IL-23
BR112022010231A2 (en) 2019-12-06 2022-09-06 Ablynx Nv POLYPEPTIDES COMPRISING SINGLE VARIABLE DOMAINS OF IMMUNOGLOBULIN TARGETED TNFALFA AND OX40L
IL293548A (en) 2019-12-09 2022-08-01 Ablynx Nv Polypeptides comprising immunoglobulin single variable domains targeting il-13 and tslp
GB201918279D0 (en) 2019-12-12 2020-01-29 Vib Vzw Glycosylated single chain immunoglobulin domains
EP4077372A1 (en) 2019-12-20 2022-10-26 Vib Vzw Nanobody exchange chromatography
WO2021140205A1 (en) 2020-01-10 2021-07-15 Confo Therapeutics N.V. Methods for generating antibodies and antibody fragments and libraries comprising same
WO2021156490A2 (en) 2020-02-06 2021-08-12 Vib Vzw Corona virus binders
KR20220144841A (en) 2020-02-21 2022-10-27 하푼 테라퓨틱스, 인크. FLT3 Binding Proteins and Methods of Use
US20230087785A1 (en) 2020-02-25 2023-03-23 Vib Vzw Leucine-Rich Repeat Kinase 2 Allosteric Modulators
CN115485289A (en) 2020-03-11 2022-12-16 宾夕法尼亚大学董事会 Methods and compositions for gene delivery using engineered viral particles
CN113461824A (en) 2020-03-31 2021-10-01 普米斯生物技术(珠海)有限公司 Platform for constructing multispecific antibody
CA3178461A1 (en) 2020-03-31 2021-10-07 Biotalys NV Anti-fungal polypeptides
WO2021213435A1 (en) 2020-04-22 2021-10-28 迈威(上海)生物科技股份有限公司 Single variable domain antibody targeting human programmed death ligand 1 (pd-l1) and derivative thereof
WO2021229104A1 (en) 2020-05-15 2021-11-18 Université de Liège Anti-cd38 single-domain antibodies in disease monitoring and treatment
KR20230012559A (en) 2020-05-19 2023-01-26 베링거 인겔하임 인터내셔날 게엠베하 Binding Molecules for Cancer Treatment
GB202101299D0 (en) 2020-06-09 2021-03-17 Avacta Life Sciences Ltd Diagnostic polypetides and methods
WO2022003156A1 (en) 2020-07-02 2022-01-06 Oncurious Nv Ccr8 non-blocking binders
EP4189061A1 (en) 2020-07-31 2023-06-07 Biotalys NV Methods of increasing recombinant protein yields
EP4216943A1 (en) 2020-09-24 2023-08-02 Vib Vzw Combination of p2y6 inhibitors and immune checkpoint inhibitors
WO2022063957A1 (en) 2020-09-24 2022-03-31 Vib Vzw Biomarker for anti-tumor therapy
EP4217390A1 (en) 2020-09-25 2023-08-02 Ablynx N.V. Polypeptides comprising immunoglobulin single variable domains targeting il-13 and ox40l
US20220119536A1 (en) 2020-10-21 2022-04-21 Boehringer Ingelheim International Gmbh Agonistic trkb binding molecules for the treatment of eye diseases
WO2022098743A1 (en) 2020-11-03 2022-05-12 Indi Molecular, Inc. Compositions, imaging, and therapeutic methods targeting folate receptor 1 (folr1)
WO2022098745A1 (en) 2020-11-03 2022-05-12 Indi Molecular, Inc. Compositions, delivery systems, and methods useful in tumor therapy
EP4255929A2 (en) 2020-12-02 2023-10-11 Vib Vzw An ltbr agonist in combination therapy against cancer
WO2022117569A1 (en) 2020-12-02 2022-06-09 Oncurious Nv A ccr8 antagonist antibody in combination with a lymphotoxin beta receptor agonist antibody in therapy against cancer
WO2022129637A1 (en) 2020-12-18 2022-06-23 Ablynx Nv T cell recruiting polypeptides based on tcr alpha/beta reactivity
EP4263602A1 (en) 2020-12-18 2023-10-25 Ablynx N.V. Polypeptides comprising immunoglobulin single variable domains targeting il-6 and tnf-alpha
WO2022129560A1 (en) 2020-12-18 2022-06-23 Ablynx Nv Polypeptides comprising immunoglobulin single variable domains targeting glypican-3 and t cell receptor
GB202020502D0 (en) 2020-12-23 2021-02-03 Vib Vzw Antibody composistion for treatment of corona virus infection
EP4267617A1 (en) 2020-12-24 2023-11-01 Vib Vzw Human ccr8 binders
US20240052045A1 (en) 2020-12-24 2024-02-15 Vib Vzw Murine cross-reactive human ccr8 binders
EP4267618A1 (en) 2020-12-24 2023-11-01 Vib Vzw Non-blocking human ccr8 binders
CN117794566A (en) 2021-02-05 2024-03-29 Vib研究所 Sha Bei viral binding agents
IL304929A (en) 2021-02-05 2023-10-01 Vib Vzw [Be/Be Sarbecovirus binders
WO2022175392A1 (en) 2021-02-17 2022-08-25 Vib Vzw Inhibition of slc4a4 in the treatment of cancer
EP4294516A1 (en) 2021-02-19 2023-12-27 Vib Vzw Cation-independent mannose-6-phosphate receptor binders
WO2022199804A1 (en) 2021-03-24 2022-09-29 Vib Vzw Nek6 inhibition to treat als and ftd
WO2022234003A1 (en) 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
WO2022242892A1 (en) 2021-05-17 2022-11-24 Université de Liège Anti-cd38 single-domain antibodies in disease monitoring and treatment
US20220411533A1 (en) 2021-06-17 2022-12-29 Boehringer Ingelheim International Gmbh Novel Tri-specific Binding Molecules
CA3225194A1 (en) 2021-06-23 2022-12-29 Vib Vzw Means and methods for selection of specific binders
CN117580865A (en) 2021-06-29 2024-02-20 山东先声生物制药有限公司 CD16 antibodies and uses thereof
CA3227972A1 (en) 2021-07-30 2023-02-02 Shandong Simcere Biopharmaceutical Co., Ltd. Anti-pvrig/anti-tigit bispecific antibodies and applications thereof
CA3228014A1 (en) 2021-07-30 2023-02-16 Vib Vzm Cation-independent mannose-6-phosphate receptor binders for targeted protein degradation
WO2023057601A1 (en) 2021-10-06 2023-04-13 Biotalys NV Anti-fungal polypeptides
TW202332694A (en) 2021-10-07 2023-08-16 英商阿凡克塔生命科學公司 Serum half-life extended pd-l1 binding polypeptides
TW202334196A (en) 2021-10-07 2023-09-01 英商阿法克塔生命科學有限公司 Pd-l1 binding polypeptides
WO2023114884A2 (en) 2021-12-15 2023-06-22 Interius Biotherapeutics, Inc. Pseudotyped viral particles, compositions comprising the same, and uses thereof
TW202342508A (en) 2021-12-17 2023-11-01 比利時商艾伯霖克斯公司 POLYPEPTIDES COMPRISING IMMUNOGLOBULIN SINGLE VARIABLE DOMAINS TARGETING TCRαβ, CD33 and CD123
US20230257448A1 (en) * 2022-01-03 2023-08-17 Twist Bioscience Corporation Bispecific sars-cov-2 antibodies and methods of use
WO2023135198A1 (en) 2022-01-12 2023-07-20 Vib Vzw Human ntcp binders for therapeutic use and liver-specific targeted delivery
WO2023148291A1 (en) 2022-02-02 2023-08-10 Biotalys NV Methods for genome editing
WO2023148397A1 (en) 2022-02-07 2023-08-10 Vib Vzw Engineered stabilizing aglycosylated fc-regions
WO2023198848A1 (en) 2022-04-13 2023-10-19 Vib Vzw An ltbr agonist in combination therapy against cancer
WO2023213751A1 (en) 2022-05-02 2023-11-09 Umc Utrecht Holding B.V Single domain antibodies for the detection of plasmin-cleaved vwf
WO2023218243A1 (en) 2022-05-12 2023-11-16 Avacta Life Sciences Limited Lag-3/pd-l1 binding fusion proteins
WO2023222825A1 (en) 2022-05-18 2023-11-23 Vib Vzw Sarbecovirus spike s2 subunit binders
WO2023242247A1 (en) 2022-06-14 2023-12-21 Ablynx Nv Immunoglobulin single variable domains targeting t cell receptor
WO2024003873A1 (en) * 2022-06-30 2024-01-04 Intrexon Actobiotics Nv D/B/A Precigen Actobio Single variable domain antibodies against tumor necrosis factor-alpha
WO2024008755A1 (en) 2022-07-04 2024-01-11 Vib Vzw Blood-cerebrospinal fluid barrier crossing antibodies
US20240052065A1 (en) 2022-07-15 2024-02-15 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
WO2024023271A1 (en) 2022-07-27 2024-02-01 Ablynx Nv Polypeptides binding to a specific epitope of the neonatal fc receptor

Citations (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4608392A (en) 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4714759A (en) 1985-12-02 1987-12-22 Whitaker Jr Robert B Immunotoxin therapy of allergy
EP0294703A2 (en) 1987-06-10 1988-12-14 Dana-Farber Cancer Institute, Inc. Bifunctional antibody constructs and method for selectively destroying cell populations
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US4940782A (en) 1987-06-08 1990-07-10 G. D. Searle & Co. Monoclonal antibodies against IgE-associated determinants, hybrid cell lines producing these antibodies, and use therefore
US4946788A (en) 1985-06-11 1990-08-07 Ciba-Geigy Corporation Purified immunoglobulin-related factor, novel monoclonal antibodies, hybridoma cell lines, processes and applications
US4962035A (en) 1987-12-01 1990-10-09 President And Fellows Of Harvard College DNA encoding IgE receptor alpha-subunit or fragment thereof
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US5091313A (en) 1988-08-05 1992-02-25 Tanox Biosystems, Inc. Antigenic epitopes of IgE present on B cell but not basophil surface
WO1993004173A1 (en) 1991-08-14 1993-03-04 Genentech, Inc. Immunoglobulin variants for specific fc epsilon receptors
US5231026A (en) 1987-12-31 1993-07-27 Tanox Biosystems, Inc. DNA encoding murine-human chimeric antibodies specific for antigenic epitopes of IgE present on the extracellular segment of the membrane domain of membrane-bound IgE
US5252467A (en) 1987-12-31 1993-10-12 Tanox Biosystems, Inc. Method of making antibodies to antigenic epitopes of IGE present on B cells but not basophil cell surface or secreted, soluble IGE
WO1994004678A1 (en) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulins devoid of light chains
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5422258A (en) 1987-12-31 1995-06-06 Tanox Biosystems, Inc. Methods for producing high affinity anti-human IgE-monoclonal antibodies which binds to IgE on IgEabearing B cells but not basophils
US5428133A (en) 1987-12-31 1995-06-27 Tanox Biosystems, Inc. Chimeric anti-human IgE-monoclonal antibody which binds to secreted IgE and membrane-bound IgE expressed by IgE-expressing B cells but notto IgE bound to FC receptors on basophils
EP0684814A1 (en) 1993-02-22 1995-12-06 Alza Corporation Compositions for oral delivery of active agents
EP0707473A1 (en) 1993-06-03 1996-04-24 Biotechnology and Biological Sciences Research Council Oral pharmaceutical compositions comprising a protein or peptide, an antibody and polymeric beads
WO1996034103A1 (en) 1995-04-25 1996-10-31 Vrije Universiteit Brussel Variable fragments of immunoglobulins - use for therapeutic or veterinary purposes
US5629001A (en) 1991-06-21 1997-05-13 University Of Cincinnati Oral administration of therapeutic proteins for treatment of infectious disease
WO1997049805A2 (en) 1996-06-27 1997-12-31 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Recognition molecules interacting specifically with the active site or cleft of a target molecule
EP0841946A1 (en) 1995-07-27 1998-05-20 Genentech, Inc. Methods for treatment of allergic asthma
WO2000023471A2 (en) 1998-10-20 2000-04-27 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Use of a cytokine-producing lactococcus strain to treat colitis
US6190662B1 (en) 1995-09-07 2001-02-20 Vlaams Interuniversitair Instituut Voor Biotechnologie (Vib) Vzw Materials and methods relating to the attachment and display of substances on cell surfaces
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6361938B1 (en) 1996-11-08 2002-03-26 Elan Corporation, Plc Peptides which enhance transport across tissues and methods of identifying and using the same
US6514496B1 (en) 1995-04-14 2003-02-04 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
WO2003035694A2 (en) 2001-10-24 2003-05-01 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
WO2003054016A2 (en) 2001-12-21 2003-07-03 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Method for cloning of variable domain sequences

Family Cites Families (141)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US460167A (en) 1891-09-29 Car-mover
SE307996B (en) 1965-11-30 1969-01-27 Asea Ab
CU22545A1 (en) * 1994-11-18 1999-03-31 Centro Inmunologia Molecular OBTAINING A CHEMICAL AND HUMANIZED ANTIBODY AGAINST THE RECEPTOR OF THE EPIDERMAL GROWTH FACTOR FOR DIAGNOSTIC AND THERAPEUTIC USE
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4405306A (en) * 1981-12-08 1983-09-20 Beecham Inc. Medicated disposable douche product
EP0095221B1 (en) 1982-05-26 1986-07-30 Agfa-Gevaert N.V. Apparatus for transferring xerographic images
GB2148299B (en) 1983-09-01 1988-01-06 Hybritech Inc Antibody compositions of therapeutic agents having an extended serum half-life
US5672347A (en) 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
JPS62175426A (en) 1986-01-27 1987-08-01 Kazufumi Shimizu Antibody and spraying agent containing said substance as active component
JP2647427B2 (en) 1987-04-24 1997-08-27 帝人株式会社 Detection method, monoclonal antibody, and detection kit for determining disease state of subject
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
DE122006000007I1 (en) 1987-12-31 2006-04-27 Tanox Biosystems Inc Antigen epitopes that are exclusively on ige-bearing B lymphocytes.
JPH01268645A (en) 1988-04-18 1989-10-26 Teijin Ltd Agent for suppressing schwartzman reaction
US5770198A (en) * 1988-05-18 1998-06-23 The Research Foundation Of The State Of New York Platelet-specific chimeric 7E3 immunoglobulin
EP0366043B1 (en) 1988-10-24 1994-03-30 Otsuka Pharmaceutical Co., Ltd. Monoclonal antibody
JP2919890B2 (en) 1988-11-11 1999-07-19 メディカル リサーチ カウンスル Single domain ligand, receptor consisting of the ligand, method for producing the same, and use of the ligand and the receptor
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
GB8905400D0 (en) * 1989-03-09 1989-04-19 Jonker Margreet Medicaments
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
SE509359C2 (en) 1989-08-01 1999-01-18 Cemu Bioteknik Ab Use of stabilized protein or peptide conjugates for the preparation of a drug
US6267964B1 (en) 1989-08-01 2001-07-31 Affibody Technology Sweden Ab Stabilized protein or peptide conjugates able to bond albumin having extended biological half-lives
US5644034A (en) * 1989-08-07 1997-07-01 Peptide Technology Ltd. Tumour necrosis factor binding ligands
US6498237B2 (en) 1989-08-07 2002-12-24 Peptech Limited Tumor necrosis factor antibodies
CA2064915C (en) 1989-08-07 2001-05-29 Deborah A. Rathjen Tumour necrosis factor binding ligands
GB8921123D0 (en) * 1989-09-19 1989-11-08 Millar Ann B Treatment of ards
US5196193A (en) * 1989-10-31 1993-03-23 Ophidian Pharmaceuticals, Inc. Antivenoms and methods for making antivenoms
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
GB9012995D0 (en) 1990-06-11 1990-08-01 Celltech Ltd Multivalent antigen-binding proteins
GB9016299D0 (en) 1990-07-25 1990-09-12 Brien Caroline J O Binding substances
JP2988635B2 (en) 1990-09-18 1999-12-13 塩野義製薬株式会社 Monoclonal antibody against human IgE
US5612034A (en) 1990-10-03 1997-03-18 Redcell, Inc. Super-globuling for in vivo extended lifetimes
US5843440A (en) * 1990-10-03 1998-12-01 Redcell Canada, Inc. Cellular and serum protein anchors for modulating pharmacokinetics
AU8869291A (en) * 1990-10-04 1992-04-28 University Of Virginia Alumni Patents Foundation, The Primate erythrocyte bound monoclonal antibody heteropolymers
US5656272A (en) 1991-03-18 1997-08-12 New York University Medical Center Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies
JP3464215B2 (en) 1991-03-21 2003-11-05 マシモ・コーポレイション Low noise optical probe
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
ATE249840T1 (en) 1991-12-13 2003-10-15 Xoma Corp METHOD AND MATERIALS FOR PRODUCING MODIFIED VARIABLE ANTIBODY DOMAIN AND THERAPEUTIC USE THEREOF
US5869619A (en) 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
EP0584421A1 (en) 1992-08-21 1994-03-02 Cécile Casterman Immunoglobulins devoid of light chains
US6765087B1 (en) * 1992-08-21 2004-07-20 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
DK0585705T3 (en) 1992-08-28 1999-07-26 Bayer Ag Use of monoclonal anti-TNF antibodies to treat bacterial meningitis
DK0589840T3 (en) 1992-09-24 2004-07-26 Novartis Ag Converted monoclonal antibodies to an immunoglobulin isotype
US6066718A (en) * 1992-09-25 2000-05-23 Novartis Corporation Reshaped monoclonal antibodies against an immunoglobulin isotype
CA2146647C (en) 1992-10-08 2009-05-05 Marc Feldmann Treatment of autoimmune and inflammatory disorders
GB9221657D0 (en) 1992-10-15 1992-11-25 Scotgen Ltd Recombinant bispecific antibodies
JPH07508763A (en) * 1992-11-20 1995-09-28 シェリング・コーポレーション Antagonist of human gamma interferon
DK0672142T3 (en) 1992-12-04 2001-06-18 Medical Res Council Multivalent and multi-specific binding proteins as well as their preparation and use
ES2126094T3 (en) 1992-12-29 1999-03-16 Genentech Inc TREATMENT OF INFLAMMATORY DISEASE OF THE INTESTINES WITH INTERFERON-GAMMA INHIBITORS.
US5888511A (en) 1993-02-26 1999-03-30 Advanced Biotherapy Concepts, Inc. Treatment of autoimmune diseases, including AIDS
EP0614984B2 (en) 1993-03-05 2010-11-03 Bayer HealthCare LLC Anti-TNF alpha human monoclonal antibodies
EP0698097B1 (en) 1993-04-29 2001-08-16 Unilever N.V. Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae
ES2162863T3 (en) * 1993-04-29 2002-01-16 Unilever Nv PRODUCTION OF ANTIBODIES OR FRAGMENTS (FUNCTIONALIZED) OF THE SAME DERIVED FROM HEAVY CHAIN IMMUNOGLOBULINS OF CAMELIDAE.
AU1441395A (en) 1993-12-21 1995-07-10 St. Louis University Ocular diagnostics and therapies
ATE164079T1 (en) * 1994-01-18 1998-04-15 Genentech Inc METHOD FOR TREATING PARASITIC INFECTIONS USING IGE ANTAGONISTS
US5747148A (en) 1994-09-12 1998-05-05 Minnesota Mining And Manufacturing Company Ink jet printing sheet
ES2197210T3 (en) * 1994-11-30 2004-01-01 Ajinomoto Co., Inc. ANTITHROMBOTIC AGENT AND MONOCLONAL ANTIBODIES AGAINST THE VON WILLEBRAND FACTOR.
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
US6410690B1 (en) * 1995-06-07 2002-06-25 Medarex, Inc. Therapeutic compounds comprised of anti-Fc receptor antibodies
CA2222231A1 (en) * 1995-06-07 1996-12-19 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth of tumors
US5637038A (en) 1995-07-10 1997-06-10 Davis; James F. Apparatus and method for skinning poultry
US20040197326A1 (en) * 1995-07-27 2004-10-07 Genentech, Inc. Method for treatment of allergic asthma
PT853487E (en) * 1995-09-18 2000-11-30 Intracel Corp NEUTRALIZING MONOCLONAL ANTIBODIES FOR RESPIRATORY SYNCYCLE VIRUS
US7368111B2 (en) * 1995-10-06 2008-05-06 Cambridge Antibody Technology Limited Human antibodies specific for TGFβ2
RU2270030C2 (en) * 1996-02-09 2006-02-20 Абботт Байотекнолоджи эЛтиди. METHOD AND OBTAINED HUMAN ANTIBODY OR ITS ANTIGEN-BINDING FRAGMENT FOR INHIBITING HUMAN TNFα ACTIVITY, APPLYING THE OBTAINED HUMAN ANTIBODY OR ITS ANTIGEN-BINDING FRAGMENT AS INGREDIENT FOR PRODUCING MEDICAMENT
US6417337B1 (en) * 1996-10-31 2002-07-09 The Dow Chemical Company High affinity humanized anti-CEA monoclonal antibodies
IT1289608B1 (en) 1997-02-05 1998-10-15 Angelini Ricerche Spa COMPOSITION FOR THERAPEUTIC OR DIAGNOSTIC USE ADMINISTRABLE VIA INTRANASAL, SUBLINGUAL OR VAGINAL
US5942602A (en) * 1997-02-13 1999-08-24 Schering Aktiengessellschaft Growth factor receptor antibodies
AUPO561697A0 (en) 1997-03-13 1997-04-10 Crc For Cardiac Technology Haemocompatible surfaces
US5994511A (en) * 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
DE29712318U1 (en) 1997-07-07 1997-10-02 Arnold Guenter Paring knife
WO1999009055A2 (en) * 1997-08-18 1999-02-25 Innogenetics N.V. Interferon-gamma-binding molecules for treating septic shock, cachexia, immune diseases and skin disorders
DE69841562D1 (en) * 1997-10-27 2010-04-29 Bac Ip Bv MULTIVALENT ANTIGENBINDING PROTEINS
US20020076404A1 (en) * 1998-01-29 2002-06-20 Chang Tse Wen Treating atopic dermatitis with IgE antagonists
EP0954978B1 (en) 1998-03-12 2011-11-30 VHsquared Limited New products comprising inactivated yeasts or moulds provided with active antibodies
HUP9900956A2 (en) * 1998-04-09 2002-04-29 Aventis Pharma Deutschland Gmbh. Single-chain multiple antigen-binding molecules, their preparation and use
US6504013B1 (en) * 2000-02-01 2003-01-07 Idexx Laboratories, Inc. Canine allergy therapeutic recombinant chimeric anti-IgE monoclonal antibody
EP1085906B1 (en) * 1998-06-10 2008-08-13 Ophidian Pharmaceuticals, Inc. Anti il6 antibodies in the prevention and treatment of inflammatory bowel disease
US6872538B1 (en) 1998-07-06 2005-03-29 Euroscreen S.A. High-throughput screening diagnostic and/or dosage method of an agonist and/or an antagonist for a calcium-coupled receptor
US6149934A (en) * 1999-04-23 2000-11-21 Kimberly-Clark Worldwide, Inc. Absorbent article having a lotionized bodyside liner
EP1124856A1 (en) * 1998-10-23 2001-08-22 The Brigham And Women's Hospital, Inc. Conformation-specific anti-von willebrand factor antibodies
EP1002861A1 (en) 1998-10-26 2000-05-24 Unilever Plc Antigen-binding proteins comprising a linker which confers restricted conformational flexibility
GB9824632D0 (en) 1998-11-10 1999-01-06 Celltech Therapeutics Ltd Biological compounds
IL127127A0 (en) 1998-11-18 1999-09-22 Peptor Ltd Small functional units of antibody heavy chain variable regions
DE19852800C1 (en) 1998-11-16 2000-04-13 Univ Albert Ludwigs Freiburg Production of antibodies to a polypeptide encoded by a known DNA sequence comprises binding of antibodies produced by DNA vaccination to immobilized recombinantly expressed polypeptide
US6329511B1 (en) 1998-12-01 2001-12-11 Protein Design Labs, Inc. Humanized antibodies to γ-interferon
US6773916B1 (en) 1999-01-05 2004-08-10 The Flinders University Of South Australia Agents and methods for treatment and diagnosis of ocular disorders
US6419934B1 (en) * 1999-02-24 2002-07-16 Edward L. Tobinick TNF modulators for treating neurological disorders associated with viral infection
CA2669512A1 (en) 1999-03-25 2000-09-28 Abbott Gmbh & Co. Kg Human antibodies that bind human il-12 and methods for producing
CN1252264C (en) 1999-04-22 2006-04-19 荷兰联合利华有限公司 Inhibition of viral infection using monovalent antigen-binding proteins
US6436401B1 (en) * 1999-09-14 2002-08-20 Milkhaus Laboratory, Inc. Methods for alleviating symptoms associated with diabetes and diabetic neuropathy comprising administration of low levels of antibodies
WO2001019871A2 (en) 1999-09-16 2001-03-22 Unilever Plc Delivery system for antidandruff agent
EP1118669A3 (en) 1999-12-17 2001-08-29 Unilever Plc Production of camelid antibodies in plants
US7608681B2 (en) 1999-12-24 2009-10-27 Genentech, Inc. Methods and compositions for prolonging elimination half-times of bioactive compounds
JP2004500086A (en) 2000-02-10 2004-01-08 アボット・ラボラトリーズ Antibodies that bind to human interleukin 18, and methods for their preparation and use
DE60138333D1 (en) 2000-03-14 2009-05-28 Unilever Nv Variable heavy chain domains of an antibody to human nutritional lipases and their uses
US7097840B2 (en) * 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
EP1284752A4 (en) * 2000-04-26 2004-08-18 Elusys Therapeutics Inc Bispecific molecules and uses thereof
AU2001264747A1 (en) 2000-05-22 2001-12-03 Idec Pharmaceuticals Corporation Identification of unique binding interactions between certain antibodies and thehuman b7.1 and b7.2 co-stimulatory antigens
EP1328626B1 (en) * 2000-05-26 2013-04-17 National Research Council Of Canada Single-domain brain-targeting antibody fragments derived from llama antibodies
US6849259B2 (en) * 2000-06-16 2005-02-01 Symphogen A/S Polyclonal antibody composition for treating allergy
US6902734B2 (en) * 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
EP1967206B1 (en) * 2000-08-30 2012-10-10 Pfizer Products Inc. Anti-IgE vaccines
WO2002020615A2 (en) 2000-09-08 2002-03-14 Micromet Ag Antibody and/or chemokine constructs which bind to a chemokine receptor and their use in immunological disorders
US6699473B2 (en) * 2000-10-13 2004-03-02 Uab Research Foundation Human anti-epidermal growth factor receptor single-chain antibodies
WO2002039121A2 (en) * 2000-11-03 2002-05-16 Board Of Regents, The University Of Texas System Methods for detecting the efficacy of anticancer treatments
ATE513854T1 (en) 2000-12-13 2011-07-15 Bac Ip B V PROTEIN GRID OF VARIABLE DOMAIN OF THE CAMEL IMMUNOGLOBULIN HEAVY CHAIN
GB0031448D0 (en) * 2000-12-22 2001-02-07 Leuven K U Res & Dev Inhibition of the vWF-collagen interaction by anti-human vWF monoclonal antibody (82D6A3) results in abolition of in vivo arterial platelet thrombus formation
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
AU2002305159B2 (en) * 2001-03-28 2007-12-13 Heska Corporation Methods of detecting early renal disease in animals
IL158126A0 (en) * 2001-03-29 2004-03-28 Univ Ramot Peptides and antibodies to muc 1 proteins
US8178098B2 (en) * 2001-04-03 2012-05-15 National Jewish Health Method to inhibit airway hyperresponsiveness using aerosolized T cell receptor antibodies
JP4398644B2 (en) * 2001-04-06 2010-01-13 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルベニア ErbB interfacial peptide mimics and methods of use thereof
PT2163256E (en) * 2001-05-11 2015-11-20 Ludwig Inst For Cancer Res Ltd Specific binding proteins and uses thereof
AU2002319402B2 (en) * 2001-06-28 2008-09-11 Domantis Limited Dual-specific ligand and its use
US7084257B2 (en) * 2001-10-05 2006-08-01 Amgen Inc. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
JP2005526769A (en) 2002-03-15 2005-09-08 ザ・ブリガーム・アンド・ウーメンズ・ホスピタル・インコーポレーテッド Central airway administration for systemic delivery of therapeutic agents
US20040063912A1 (en) 2002-03-15 2004-04-01 The Brigham And Women's Hospital, Inc. Central airway administration for systemic delivery of therapeutics
CN1678634A (en) * 2002-06-28 2005-10-05 多曼蒂斯有限公司 Immunoglobulin single variable antigen combination area and its opposite constituent
EP1546203B1 (en) * 2002-08-01 2012-06-20 Immunomedics, Inc. Alpha-fetoprotein immu31 antibodies and fusion proteins and methods of use thereof
US20100003253A1 (en) * 2002-11-08 2010-01-07 Ablynx N.V. Single domain antibodies directed against epidermal growth factor receptor and uses therefor
US20060034845A1 (en) * 2002-11-08 2006-02-16 Karen Silence Single domain antibodies directed against tumor necrosis factor alpha and uses therefor
US20060228355A1 (en) * 2003-11-07 2006-10-12 Toon Laeremans Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
EP2336179A1 (en) * 2002-11-08 2011-06-22 Ablynx N.V. Stabilized single domain antibodies
AU2003286004A1 (en) 2002-11-08 2004-06-07 Ablynx N.V. Single domain antibodies directed against interferon- gamma and uses therefor
US20060034833A1 (en) * 2002-11-08 2006-02-16 Els Beirnaert Single domain antibodies directed against interferron-gamma and uses therefor
ES2466716T3 (en) * 2002-11-08 2014-06-11 Ablynx N.V. Stabilized single domain antibodies
US9320792B2 (en) * 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
PT1639011E (en) * 2003-06-30 2009-01-20 Domantis Ltd Pegylated single domain antibodies (dab)
EP1735348B1 (en) * 2004-03-19 2012-06-20 Imclone LLC Human anti-epidermal growth factor receptor antibody
US8105592B2 (en) 2004-11-25 2012-01-31 Vhsquared Limited Heavy chain and single domain antibodies
JP2008521870A (en) 2004-12-02 2008-06-26 ドマンティス リミテッド Anti-IL-1R1 single domain antibody and therapeutic use
CN103254309B (en) 2005-05-18 2017-09-26 埃博灵克斯股份有限公司 For the improved nanometer body of tumor necrosis factor αTM
DE102005023617A1 (en) 2005-05-21 2006-11-23 Aspre Ag Method for mixing colors in a display
EP1754955B1 (en) 2005-08-19 2015-05-06 Aisin Aw Co., Ltd. Navigation method and corresponding system for determining a travel related time
CN101426815A (en) 2005-12-06 2009-05-06 杜门蒂斯有限公司 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor
EA201000785A1 (en) 2007-12-13 2011-02-28 Глаксо Груп Лимитед COMPOSITIONS FOR DELIVERY IN LIGHT
WO2009127691A1 (en) 2008-04-17 2009-10-22 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same

Patent Citations (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
US4608392A (en) 1983-08-30 1986-08-26 Societe Anonyme Dite: L'oreal Method for producing a non greasy protective and emollient film on the skin
US4946788A (en) 1985-06-11 1990-08-07 Ciba-Geigy Corporation Purified immunoglobulin-related factor, novel monoclonal antibodies, hybridoma cell lines, processes and applications
US4714759A (en) 1985-12-02 1987-12-22 Whitaker Jr Robert B Immunotoxin therapy of allergy
US4940782A (en) 1987-06-08 1990-07-10 G. D. Searle & Co. Monoclonal antibodies against IgE-associated determinants, hybrid cell lines producing these antibodies, and use therefore
EP0294703A2 (en) 1987-06-10 1988-12-14 Dana-Farber Cancer Institute, Inc. Bifunctional antibody constructs and method for selectively destroying cell populations
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4962035A (en) 1987-12-01 1990-10-09 President And Fellows Of Harvard College DNA encoding IgE receptor alpha-subunit or fragment thereof
US5422258A (en) 1987-12-31 1995-06-06 Tanox Biosystems, Inc. Methods for producing high affinity anti-human IgE-monoclonal antibodies which binds to IgE on IgEabearing B cells but not basophils
US5231026A (en) 1987-12-31 1993-07-27 Tanox Biosystems, Inc. DNA encoding murine-human chimeric antibodies specific for antigenic epitopes of IgE present on the extracellular segment of the membrane domain of membrane-bound IgE
US5252467A (en) 1987-12-31 1993-10-12 Tanox Biosystems, Inc. Method of making antibodies to antigenic epitopes of IGE present on B cells but not basophil cell surface or secreted, soluble IGE
US5428133A (en) 1987-12-31 1995-06-27 Tanox Biosystems, Inc. Chimeric anti-human IgE-monoclonal antibody which binds to secreted IgE and membrane-bound IgE expressed by IgE-expressing B cells but notto IgE bound to FC receptors on basophils
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US5091313A (en) 1988-08-05 1992-02-25 Tanox Biosystems, Inc. Antigenic epitopes of IgE present on B cell but not basophil surface
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5629001A (en) 1991-06-21 1997-05-13 University Of Cincinnati Oral administration of therapeutic proteins for treatment of infectious disease
WO1993004173A1 (en) 1991-08-14 1993-03-04 Genentech, Inc. Immunoglobulin variants for specific fc epsilon receptors
WO1994004678A1 (en) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulins devoid of light chains
EP0684814A1 (en) 1993-02-22 1995-12-06 Alza Corporation Compositions for oral delivery of active agents
EP0707473A1 (en) 1993-06-03 1996-04-24 Biotechnology and Biological Sciences Research Council Oral pharmaceutical compositions comprising a protein or peptide, an antibody and polymeric beads
US6514496B1 (en) 1995-04-14 2003-02-04 Inhale Therapeutic Systems, Inc. Dispersible antibody compositions and methods for their preparation and use
WO1996034103A1 (en) 1995-04-25 1996-10-31 Vrije Universiteit Brussel Variable fragments of immunoglobulins - use for therapeutic or veterinary purposes
EP0841946A1 (en) 1995-07-27 1998-05-20 Genentech, Inc. Methods for treatment of allergic asthma
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6190662B1 (en) 1995-09-07 2001-02-20 Vlaams Interuniversitair Instituut Voor Biotechnologie (Vib) Vzw Materials and methods relating to the attachment and display of substances on cell surfaces
WO1997049805A2 (en) 1996-06-27 1997-12-31 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Recognition molecules interacting specifically with the active site or cleft of a target molecule
US6361938B1 (en) 1996-11-08 2002-03-26 Elan Corporation, Plc Peptides which enhance transport across tissues and methods of identifying and using the same
WO2000023471A2 (en) 1998-10-20 2000-04-27 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Use of a cytokine-producing lactococcus strain to treat colitis
WO2003035694A2 (en) 2001-10-24 2003-05-01 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
WO2003054016A2 (en) 2001-12-21 2003-07-03 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Method for cloning of variable domain sequences

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
BANIYASH ET AL., MOLECULAR IMMUNOLOGY, vol. 25, 1988, pages 705 - 711
BANIYASH; ESHHAR, EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 14, 1984, pages 799 - 807
BLATTLER, BIOCHEMISTRY, vol. 24, pages 1517 - 1524
CARMAN, C.V.; BENOVIC, J.L., CURRENT OPINION IN NEUROBIOLOGY, vol. 8, 1998, pages 335 - 344
CHAVEY C; MARI B; MONTHOUEL MN; BONNAFOUS S; ANGLARD P; VAN OBBERGHEN E; TARTARE-DECKERT S., J. BIOL. CHEM., vol. 278, 2003, pages 11888 - 11896
CHOMCZYNSKI P; SACCHÍ N: "Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction", ANAL BIOCHEM, vol. 162, 1987, pages 156 - 159
CHURG A; WANG RD; TAI H; WANG X; XIE C; DAI J; SHAPIRO SD; WRIGHT JL, AM. J. RESPIR. CRIT. CARE MED., vol. 167, 2003, pages 1083 - 1089
COHEN S; USHIRO H; STOSCHECK C; CHINKERS M: "A native 170,000 epidermal growth factor receptor-kinase complex from shed plasma membrane vesicles", J. BIOL. CHEM., vol. 257, 1982, pages 1523 - 31
CONRATH ET AL., J BIOI CHEM, vol. 276, 2001, pages 7346 - 7350
DE HAARD HJ; VAN NEER N; REURS A; HUFTON SE; ROOVERS RC; HENDERIKX P; DE BRUINE AP; ARENDS JW; HOOGENBOOM HR: "A large non-immunized human Fab fragment phage library that permits rapid isolation and kinetic analysis of high affinity antibodies", J. BIOI. CHEM., vol. 274, 1999, pages 18218 - 30
DEFIZE LH; MOOLENAAR WH; VAN DER SAAG PT; DE LAAT SW: "Dissociation of cellular responses to epidermal growth factor using anti-receptor monoclonal antibodies", EMBO J., vol. 5, 1986, pages 1187 - 92
DING Y; SHIMADA Y; GORRIN-RIVAS MJ; ITAMI A; LI Z; HONG T; MAEDA M; KOMOTO I; KAWABE A; KAGANOI J, ONCOLOGY, vol. 63, no. 4, 2002, pages 378 - 84
G. KOJOUHAROFF ET AL., CIIN. EXP. IMMUNOL., vol. 107, 1997, pages 353 - 8
G. KOJOUHAROFF, CLIN. EXP. IMMUNOL., vol. 107, 1997, pages 353 - 8
HOOK ET AL., FEDERATION PROCEEDINGS, vol. 40, no. 3
HOOK, FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY, 71ST ANNUAL MEETING, 1987
J. BIOI. CHEM., vol. 49, 1921, pages 183
KASSNER, BIOCHEM BIOPHYS RES COMMUN, vol. 264, 1999, pages 921 - 928
KERKELA E; ALA-AHO R; JESKANEN L; RECHARDT 0; GRENMAN R; SHAPIRO SD; KAHARI VM; SAARIALHO-KERE U, J INVEST DERMATOL, vol. 114, no. 6, June 2000 (2000-06-01), pages 1113 - 9
KOJOUHAROFF G; HANS W; OBERMEIER F; MANNEL DN; ANDUS T; SCHOLMERICH J; GROSS V; FALK W: "Neutralization of tumour necrosis factor (TNF) but not of IL-1 reduces inflammation in chronic dextran sulphate sodium-induced colitis in mice", CLIN EXP IMMUNOL, vol. 107, 1997, pages 353 - 8
LAROCCA D ET AL., CURR. PHARM. BIOTECHNOL, vol. 3, 2002, pages 45 - 57
LAROCCO, MOLECULAR THERAPY, vol. 3, 2001, pages 476 - 484
MAGNI ET AL., ANAL BIOCHEM, vol. 298, 2001, pages 181 - 188
MARTIN, E.W.: "Remington's Pharmaceutical Sciences, ed. 4", MACK PUBLISHING CO.
MUYIDERMANS, S., REVIEWS IN MOLECULAR BIOTECHNOLOGY, vol. 74, 2001, pages 277 - 303
MUYLDERMANS, S., REVIEWS IN MOLECULAR BIOTECHNOLOGY, vol. 74, 2001, pages 277 - 303
OKAYASU 1; HATAKEYAMA S; YAMADA M; OHKUSA T; INAGAKI Y; NAKAYA R: "A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice", GASTROENTEROLOGY, vol. 98, 1990, pages 694 - 702
OKAYASU ET AL., GASTROENTEROLOGY, vol. 98, 1990, pages 694 - 702
PARTRIDGE, ADV. DRUG DELIVERY REVIEWS, vol. 15, 1995, pages 5 - 36
POUL MA ET AL., J MOL BIOL, vol. 288, 1999, pages 203 - 211
POUL MA ET AL., J MOL BIOL, vol. 301, 2000, pages 1149 - 1161
POUL MA, J MOL BIOL, vol. 288, 1999, pages 203 - 211
POUL MA; BECERRIL B; NIELSEN UB; MORISSON P; MARKS JD: "Selection of tumor-specific internalizing human antibodies from phage libraries", J. MOL, BIOL., vol. 301, 2000, pages 1149 - 61
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623 - 2832
R LANG; A KOCOUREK; M BRAUN; H TSCHESCHE; R HUBER; W BODE; K MASKOS, J MOL BIOL, vol. 312, no. 4, September 2001 (2001-09-01), pages 731 - 42
RUP; KAHN, INTERNATIONAL ARCHIVES ALLERGY AND APPLIED IMMUNOLOGY, vol. 89, 1989, pages 387 - 393
SALMELA MT; PENDER SL; REUNALA T; MACDONALD T; SAARIALHO-KERE U., GUT, vol. 48, no. 4, 2001, pages 496 - 502
SAMBROOK ET AL.: "Molecular Cloning, Laboratory Manuel", COLD SPRING, HARBOR LABORATORY PRESS
SATO JD; KAWAMOTO T; LE AD; MENDELSOHN J; POLIKOFF J; SATO GH: "Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors", MOL. BIO!. MED., vol. 1, 1983, pages 511 - 529
THOMPSON, J.D.; HIGGINS, D.G.; GIBSON, T.J.: "CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position specific gap penalties and weight matrix choice", NUCLEIC ACIDS RESEARCH, June 1994 (1994-06-01)
YOSHIKATSU KANEKO; MINORU SAKATSUME; YUANSHENG XIE; TAKESHI KURODA; MICHIKO LGASHIMA; ICHIEI NARITA; FUMITAKE GEJYO, THE JOURNAL OF IMMUNOLOGY, vol. 170, 2003, pages 3377 - 3385

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2530553C2 (en) * 2012-11-07 2014-10-10 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Московский государственный университет имени М.В. Ломоносова" (МГУ) Recombinant single-domain antibody able to bind human tumour necrosis factor specifically and its derivatives

Also Published As

Publication number Publication date
EP2316852B1 (en) 2014-03-05
JP2011256169A (en) 2011-12-22
JP2006519763A (en) 2006-08-31
WO2004041865A2 (en) 2004-05-21
NZ540194A (en) 2008-07-31
WO2004041863A3 (en) 2004-07-15
AU2010226938B2 (en) 2012-03-29
US20100003248A1 (en) 2010-01-07
RU2704232C2 (en) 2019-10-24
JP2006524036A (en) 2006-10-26
US20110184145A1 (en) 2011-07-28
WO2004041862A3 (en) 2004-06-24
AU2010226938A1 (en) 2010-10-28
EP2284192A3 (en) 2011-07-20
EP3299393A1 (en) 2018-03-28
RU2012108416A (en) 2013-09-10
EP1900753A3 (en) 2008-04-09
MXPA05004955A (en) 2005-11-17
CA2505325C (en) 2014-02-25
JP2006520584A (en) 2006-09-14
EP2258392A1 (en) 2010-12-08
CA2505326A1 (en) 2004-05-21
HK1082745A1 (en) 2006-06-16
US20100003249A1 (en) 2010-01-07
NZ540195A (en) 2009-01-31
US20090324512A1 (en) 2009-12-31
IL168357A (en) 2011-09-27
EP1900753B1 (en) 2017-08-09
JP2011024581A (en) 2011-02-10
CA2505316A1 (en) 2004-05-21
CA2505325A1 (en) 2004-05-21
EP2390268A1 (en) 2011-11-30
NZ563471A (en) 2009-04-30
EP2316852A1 (en) 2011-05-04
EP1900753A2 (en) 2008-03-19
US20100040613A1 (en) 2010-02-18
CA2505316C (en) 2014-08-05
EP2390268B1 (en) 2017-11-01
WO2004041865A3 (en) 2004-07-15
US20100021459A1 (en) 2010-01-28
WO2004041863A2 (en) 2004-05-21
EP2336179A1 (en) 2011-06-22
EP2301967A3 (en) 2011-07-13
NZ540196A (en) 2008-09-26
US9243065B2 (en) 2016-01-26
WO2004041862A2 (en) 2004-05-21
US20060115470A1 (en) 2006-06-01
EP2284192A2 (en) 2011-02-16

Similar Documents

Publication Publication Date Title
US9243065B2 (en) Polypeptide constructs including VHH directed against EGFR for intracellular delivery
AU2003283137B8 (en) Camelidae antibodies against immunoglobulin E and use thereof for the treatment of allergic disorders
US9725522B2 (en) Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US20110123529A1 (en) Single domain antibodies directed against epidermal growth factor receptor and uses therefor
EP1687338B1 (en) Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses thereof
US20070178082A1 (en) Stabilized single domain antibodies

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20100922

AC Divisional application: reference to earlier application

Ref document number: 1558650

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Free format text: PREVIOUS MAIN CLASS: C07K0016180000

Ipc: C07K0016420000

PUAL Search report despatched

Free format text: ORIGINAL CODE: 0009013

AK Designated contracting states

Kind code of ref document: A3

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 31/16 20060101ALI20110607BHEP

Ipc: A61P 31/06 20060101ALI20110607BHEP

Ipc: A61P 35/00 20060101ALI20110607BHEP

Ipc: A61P 11/06 20060101ALI20110607BHEP

Ipc: A61K 39/395 20060101ALI20110607BHEP

Ipc: C12N 15/13 20060101ALI20110607BHEP

Ipc: C07K 16/28 20060101ALI20110607BHEP

Ipc: C07K 16/40 20060101ALI20110607BHEP

Ipc: C07K 16/10 20060101ALI20110607BHEP

Ipc: C07K 16/12 20060101ALI20110607BHEP

Ipc: C07K 16/30 20060101ALI20110607BHEP

Ipc: C07K 16/24 20060101ALI20110607BHEP

Ipc: C07K 16/42 20060101AFI20110607BHEP

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ABLYNX N.V.

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20120114